US20070015252A1 - Corynebacterium glutamicum genes encoding regulatory proteins - Google Patents

Corynebacterium glutamicum genes encoding regulatory proteins Download PDF

Info

Publication number
US20070015252A1
US20070015252A1 US11/508,066 US50806606A US2007015252A1 US 20070015252 A1 US20070015252 A1 US 20070015252A1 US 50806606 A US50806606 A US 50806606A US 2007015252 A1 US2007015252 A1 US 2007015252A1
Authority
US
United States
Prior art keywords
protein
nucleic acid
gene
acid molecule
amino acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/508,066
Inventor
Markus Pompejus
Burkhard Kroger
Hartwig Schroder
Oskar Zelder
Gregor Haberhauer
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
BASF SE
Original Assignee
BASF SE
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by BASF SE filed Critical BASF SE
Priority to US11/508,066 priority Critical patent/US20070015252A1/en
Assigned to BASF AG reassignment BASF AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HABERHAUER, GREGOR, KROGER, BURKHARD, POMPEJUS, MARKUS, SCHRODER, HARTWIG, ZELDER, OSKAR
Publication of US20070015252A1 publication Critical patent/US20070015252A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P7/00Preparation of oxygen-containing organic compounds
    • C12P7/64Fats; Fatty oils; Ester-type waxes; Higher fatty acids, i.e. having at least seven carbon atoms in an unbroken chain bound to a carboxyl group; Oxidised oils or fats
    • C12P7/6436Fatty acid esters
    • C12P7/6445Glycerides
    • C12P7/6472Glycerides containing polyunsaturated fatty acid [PUFA] residues, i.e. having two or more double bonds in their backbone
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/34Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Corynebacterium (G)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/18Carboxylic ester hydrolases (3.1.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/90Isomerases (5.)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P13/00Preparation of nitrogen-containing organic compounds
    • C12P13/04Alpha- or beta- amino acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P17/00Preparation of heterocyclic carbon compounds with only O, N, S, Se or Te as ring hetero atoms
    • C12P17/10Nitrogen as only ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P7/00Preparation of oxygen-containing organic compounds
    • C12P7/64Fats; Fatty oils; Ester-type waxes; Higher fatty acids, i.e. having at least seven carbon atoms in an unbroken chain bound to a carboxyl group; Oxidised oils or fats
    • C12P7/6409Fatty acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P7/00Preparation of oxygen-containing organic compounds
    • C12P7/64Fats; Fatty oils; Ester-type waxes; Higher fatty acids, i.e. having at least seven carbon atoms in an unbroken chain bound to a carboxyl group; Oxidised oils or fats
    • C12P7/6409Fatty acids
    • C12P7/6427Polyunsaturated fatty acids [PUFA], i.e. having two or more double bonds in their backbone
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P7/00Preparation of oxygen-containing organic compounds
    • C12P7/64Fats; Fatty oils; Ester-type waxes; Higher fatty acids, i.e. having at least seven carbon atoms in an unbroken chain bound to a carboxyl group; Oxidised oils or fats
    • C12P7/6436Fatty acid esters
    • C12P7/6445Glycerides
    • C12P7/6463Glycerides obtained from glyceride producing microorganisms, e.g. single cell oil
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • Certain products and by-products of naturally-occurring metabolic processes in cells have utility in a wide array of industries, including the food, feed, cosmetics, and pharmaceutical industries.
  • These molecules collectively termed ‘fine chemicals’, include organic acids, both proteinogenic and non-proteinogenic amino acids, nucleotides and nucleosides, lipids and fatty acids, diols, carbohydrates, aromatic compounds, vitamins and cofactors, and enzymes.
  • Their production is most conveniently performed through the large-scale culture of bacteria developed to produce and secrete large quantities of one or more desired molecules.
  • One particularly useful organism for this purpose is Corynebacterium glutamicum , a gram positive, nonpathogenic bacterium. Through strain selection, a number of mutant strains have been developed which produce an array of desirable compounds. However, selection of strains improved for the production of a particular molecule is a time-consuming and difficult process.
  • the invention provides novel bacterial nucleic acid molecules which have a variety of uses. These uses include the identification of microorganisms which can be used to produce fine chemicals, the modulation of fine chemical production in C. glutamicum or related bacteria, the typing or identification of C. glutamicum or related bacteria, as reference points for mapping the C. glutamicum genome, and as markers for transformation. These novel nucleic acid molecules encode proteins, referred to herein as metabolic regulatory (MR) proteins.
  • MR metabolic regulatory
  • C. glutamicum is a gram positive, aerobic bacterium which is commonly used in industry for the large-scale production of a variety of fine chemicals, and also for the degradation of hydrocarbons (such as in petroleum spills) and for the oxidation of terpenoids.
  • the MR nucleic acid molecules of the invention therefore, can be used to identify microorganisms which can be used to produce fine chemicals, e.g., by fermentation processes.
  • Modulation of the expression of the MR nucleic acids of the invention, or modification of the sequence of the MR nucleic acid molecules of the invention, can be used to modulate the production of one or more fine chemicals from a microorganism (e.g., to improve the yield or production of one or more fine chemicals from a Corynebacterium or Brevibacterium species).
  • the MR nucleic acids of the invention may also be used to identify an organism as being Corynebacterium glutamicum or a close relative thereof, or to identify the presence of C. glutamicum or a relative thereof in a mixed population of microorganisms.
  • the invention provides the nucleic acid sequences of a number of C. glutamicum genes; by probing the extracted genomic DNA of a culture of a unique or mixed population of microorganisms under stringent conditions with a probe spanning a region of a C. glutamicum gene which is unique to this organism, one can ascertain whether this organism is present.
  • Corynebacterium glutamicum itself is nonpathogenic, it is related to species pathogenic in humans, such as Corynebacterium diphtheriae (the causative agent of diphtheria); the detection of such organisms is of significant clinical relevance.
  • the MR nucleic acid molecules of the invention may also serve as reference points for mapping of the C. glutamicum genome, or of genomes of related organisms. Similarly, these molecules, or variants or portions thereof, may serve as markers for genetically engineered Corynebacterium or Brevibacterium species.
  • the MR proteins encoded by the novel nucleic acid molecules of the invention are capable of, for example, performing a function involved in the transcriptional, translational, or posttranslational regulation of proteins important for the normal metabolic functioning of cells.
  • Corynebacterium glutamicum such as those disclosed in Sinskey et al., U.S. Pat. No. 4,649,119, and techniques for genetic manipulation of C. glutamicum and the related Brevibacterium species (e.g., lactofermentum ) (Yoshihama et al, J. Bacteriol. 162: 591-597 (1985); Katsumata et al., J. Bacteriol.
  • nucleic acid molecules of the invention may be utilized in the genetic engineering of this organism to make it a better or more efficient producer of one or more fine chemicals.
  • This improved yield, production and/or efficiency of production of a fine chemical may be due to a direct effect of manipulation of a gene of the invention, or it may be due to an indirect effect of such manipulation.
  • alterations in C. glutamicum MR proteins which normally regulate the yield, production and/or efficiency of production of a fine chemical metabolic pathways may have a direct impact on the overall production or rate of production of one or more of these desired compounds from this organism.
  • Alterations in the proteins involved in these metabolic pathways may also have an indirect impact on the yield, production and/or efficiency of production of a desired fine chemical. Regulation of metabolism is necessarily complex, and the regulatory mechanisms governing different pathways may intersect at multiple points such that more than one pathway can be rapidly adjusted in accordance with a particular cellular event.
  • the nucleic acid and protein molecules of the invention may be utilized to directly improve the yield, production, and/or efficiency of production of one or more desired fine chemicals from Corynebacterium glutamicum .
  • one or more of the regulatory proteins of the invention may be manipulated such that its function is modulated. For example, the mutation of an MR protein involved in the repression of transcription of a gene encoding an enzyme which is required for the biosynthesis of an amino acid such that it no longer is able to repress transcription may result in an increase in production of that amino acid.
  • the alteration of activity of an MR protein resulting in increased translation or activating posttranslational modification of a C.
  • glutamicum protein involved in the biosynthesis of a desired fine chemical may in turn increase the production of that chemical.
  • the opposite situation may also be of benefit: by increasing the repression of transcription or translation, or by posttranslational negative modification of a C. glutamicum protein involved in the regulation of a degradative pathway for a compound, one may increase the production of this chemical. In each case, the overall yield or rate of production of the desired fine chemical may be increased.
  • alterations in the protein and nucleotide molecules of the invention may improve the yield, production, and/or efficiency of production of fine chemicals through indirect mechanisms.
  • the metabolism of any one compound is necessarily intertwined with other biosynthetic and degradative pathways within the cell, and necessary cofactors, intermediates, or substrates in one pathway are likely supplied or limited by another such pathway. Therefore, by modulating the activity of one or more of the regulatory proteins of the invention, the production or efficiency of activity of another fine chemical biosynthetic or degradative pathway may be impacted.
  • the manipulation of one or more regulatory proteins may increase the overall ability of the cell to grow and multiply in culture, particularly in large-scale fermentative culture, where growth conditions may be suboptimal.
  • an MR protein of the invention which would normally cause a repression in the biosynthesis of nucleotides in response to suboptimal extracellular supplies of nutrients (thereby preventing cell division) such that it is decreased in repressor ability, one may increase the biosynthesis of nucleotides and perhaps increase cell division.
  • Changes in MR proteins which result in increased cell growth and division in culture may result in an increase in yield, production, and/or efficiency of production of one or more desired fine chemicals from the culture, due at least to the increased number of cells producing the chemical in the culture.
  • the invention provides novel nucleic acid molecules which encode proteins, referred to herein as metabolic pathway proteins (MR), which are capable of, for example, performing an enzymatic step involved in the transcriptional, translational, or posttranslational regulation of metabolic pathways in C. glutamicum .
  • MR metabolic pathway proteins
  • Nucleic acid molecules encoding an MR protein are referred to herein as MR nucleic acid molecules.
  • the MR protein participates in the transcriptional, translational, or posttranslational regulation of one or more metabolic pathways. Examples of such proteins include those encoded by the genes set forth in Table 1.
  • one aspect of the invention pertains to isolated nucleic acid molecules (e.g., cDNAs, DNAs, or RNAs) comprising a nucleotide sequence encoding an MR protein or biologically active portions thereof, as well as nucleic acid fragments suitable as primers or hybridization probes for the detection or amplification of MR-encoding nucleic acid (e.g., DNA or mRNA).
  • the isolated nucleic acid molecule comprises one of the nucleotide sequences set forth in Appendix A or the coding region or a complement thereof of one of these nucleotide sequences.
  • the isolated nucleic acid molecule of the invention comprises a nucleotide sequence which hybridizes to or is at least about 50%, preferably at least about 60%, more preferably at least about 70%, 80% or 90%, and even more preferably at least about 95%, 96%, 97%, 98%, 99% or more homologous to a nucleotide sequence set forth in Appendix A, or a portion thereof.
  • the isolated nucleic acid molecule encodes one of the amino acid sequences set forth in Appendix B.
  • the preferred MR proteins of the present invention also preferably possess at least one of the MR activities described herein.
  • the isolated nucleic acid molecule encodes a protein or portion thereof wherein the protein or portion thereof includes an amino acid sequence which is sufficiently homologous to an amino acid sequence of Appendix B, e.g., sufficiently homologous to an amino acid sequence of Appendix B such that the protein or portion thereof maintains an MR activity.
  • the protein or portion thereof encoded by the nucleic acid molecule maintains the ability to transcriptionally, translationally, or posttranslationally regulate a metabolic pathway in C. glutamicum .
  • the protein encoded by the nucleic acid molecule is at least about 50%, preferably at least about 60%, and more preferably at least about 70%, 80%, or 90% and most preferably at least about 95%, 96%, 97%, 98%, or 99% or more homologous to an amino acid sequence of Appendix B (e.g., an entire amino acid sequence selected from those sequences set forth in Appendix B).
  • the protein is a full length C. glutamicum protein which is substantially homologous to an entire amino acid sequence of Appendix B (encoded by an open reading frame shown in Appendix A).
  • the isolated nucleic acid molecule is derived from C. glutamicum and encodes a protein (e.g., an MR fusion protein) which includes a biologically active domain which is at least about 50% or more homologous to one of the amino acid sequences of Appendix B and is able to transcriptionally, translationally, or posttranslationally regulate a metabolic pathway in C. glutamicum , or has one or more of the activities set forth in Table 1, and which also includes heterologous nucleic acid sequences encoding a heterologous polypeptide or regulatory regions.
  • a protein e.g., an MR fusion protein
  • the isolated nucleic acid molecule is at least 15 nucleotides in length and hybridizes under stringent conditions to a nucleic acid molecule comprising a nucleotide sequence of Appendix A.
  • the isolated nucleic acid molecule corresponds to a naturally-occurring nucleic acid molecule. More preferably, the isolated nucleic acid encodes a naturally-occurring C. glutamicum MR protein, or a biologically active portion thereof.
  • vectors e.g., recombinant expression vectors, containing the nucleic acid molecules of the invention, and host cells into which such vectors have been introduced.
  • a host cell is used to produce an MR protein by culturing the host cell in a suitable medium. The MR protein can be then isolated from the medium or the host cell.
  • Yet another aspect of the invention pertains to a genetically altered microorganism in which an MR gene has been introduced or altered.
  • the genome of the microorganism has been altered by introduction of a nucleic acid molecule of the invention encoding wild-type or mutated MR sequence as a transgene.
  • an endogenous MR gene within the genome of the microorganism has been altered, e.g., functionally disrupted, by homologous recombination with an altered MR gene.
  • an endogenous or introduced MR gene in a microorganism has been altered by one or more point mutations, deletions, or inversions, but still encodes a functional MR protein.
  • one or more of the regulatory regions (e.g., a promoter, repressor, or inducer) of an MR gene in a microorganism has been altered (e.g., by deletion, truncation, inversion, or point mutation) such that the expression of the MR gene is modulated.
  • the microorganism belongs to the genus Corynebacterium or Brevibacterium , with Corynebacterium glutamicum being particularly preferred.
  • the microorganism is also utilized for the production of a desired compound, such as an amino acid, with lysine being particularly preferred.
  • the invention provides a method of identifying the presence or activity of Cornyebacterium diphtheriae in a subject.
  • This method includes detection of one or more of the nucleic acid or amino acid sequences of the invention (e.g., the sequences set forth in Appendix A or Appendix B) in a subject, thereby detecting the presence or activity of Corynebacterium diphtheriae in the subject.
  • Still another aspect of the invention pertains to an isolated MR protein or a portion, e.g., a biologically active portion, thereof.
  • the isolated MR protein or portion thereof transcriptionally, translationally, or posttranslationally regulates one or more metabolic pathways in C. glutamicum .
  • the isolated MR protein or portion thereof is sufficiently homologous to an amino acid sequence of Appendix B such that the protein or portion thereof maintains the ability to transcriptionally, translationally, or posttranslationally regulate one or more metabolic pathways in C. glutamicum.
  • the invention also provides an isolated preparation of an MR protein.
  • the MR protein comprises an amino acid sequence of Appendix B.
  • the invention pertains to an isolated full length protein which is substantially homologous to an entire amino acid sequence of Appendix B (encoded by an open reading frame set forth in Appendix A).
  • the protein is at least about 50%, preferably at least about 60%, and more preferably at least about 70%, 80%, or 90%, and most preferably at least about 95%, 96%, 97%, 98%, or 99% or more homologous to an entire amino acid sequence of Appendix B.
  • the isolated MR protein comprises an amino acid sequence which is at least about 50% or more homologous to one of the amino acid sequences of Appendix B and is able to transcriptionally, translationally, or posttranslationally regulate one or more metabolic pathways in C. glutamicum , or has one or more of the activities set forth in Table 1.
  • the isolated MR protein can comprise an amino acid sequence which is encoded by a nucleotide sequence which hybridizes, e.g., hybridizes under stringent conditions, or is at least about 50%, preferably at least about 60%, more preferably at least about 70%, 80%, or 90%, and even more preferably at least about 95%, 96%, 97%, 98,%, or 99% or more homologous, to a nucleotide sequence of Appendix B. It is also preferred that the preferred forms of MR proteins also have one or more of the MR bioactivities described herein.
  • the MR polypeptide, or a biologically active portion thereof, can be operatively linked to a non-MR polypeptide to form a fusion protein.
  • this fusion protein has an activity which differs from that of the MR protein alone.
  • this fusion protein transcriptionally, translationally, or posttranslationally regulates one or more metabolic pathways in C. glutamicum .
  • integration of this fusion protein into a host cell modulates production of a desired compound from the cell.
  • the invention provides methods for screening molecules which modulate the activity of an MR protein, either by interacting with the protein itself or a substrate or binding partner of the MR protein, or by modulating the transcription or translation of an MR nucleic acid molecule of the invention.
  • Another aspect of the invention pertains to a method for producing a fine chemical. This method involves the culturing of a cell containing a vector directing the expression of an MR nucleic acid molecule of the invention, such that a fine chemical is produced.
  • this method further includes the step of obtaining a cell containing such a vector, in which a cell is transfected with a vector directing the expression of an MR nucleic acid.
  • this method further includes the step of recovering the fine chemical from the culture.
  • the cell is from the genus Corynebacterium or Brevibacterium , or is selected from those strains set forth in Table 3.
  • Another aspect of the invention pertains to methods for modulating production of a molecule from a microorganism. Such methods include contacting the cell with an agent which modulates MR protein activity or MR nucleic acid expression such that a cell associated activity is altered relative to this same activity in the absence of the agent.
  • the cell is modulated for one or more C. glutamicum metabolic pathway regulatory systems, such that the yields or rate of production of a desired fine chemical by this microorganism is improved.
  • the agent which modulates MR protein activity can be an agent which stimulates MR protein activity or MR nucleic acid expression.
  • agents which stimulate MR protein activity or MR nucleic acid expression include small molecules, active MR proteins, and nucleic acids encoding MR proteins that have been introduced into the cell.
  • agents which inhibit MR activity or expression include small molecules and antisense MR nucleic acid molecules.
  • Another aspect of the invention pertains to methods for modulating yields of a desired compound from a cell, involving the introduction of a wild-type or mutant MR gene into a cell, either maintained on a separate plasmid or integrated into the genome of the host cell. If integrated into the genome, such integration can be random, or it can take place by homologous recombination such that the native gene is replaced by the introduced copy, causing the production of the desired compound from the cell to be modulated.
  • said yields are increased.
  • said chemical is a fine chemical.
  • said fine chemical is an amino acid.
  • said amino acid is L-lysine.
  • the present invention provides MR nucleic acid and protein molecules which are involved in the regulation of metabolism in Corynebacterium glutamicum , including regulation of fine chemical metabolism.
  • the molecules of the invention may be utilized in the modulation of production of fine chemicals from microorganisms, such as C.
  • glutamicum either directly (e.g., where modulation of the activity of a lysine biosynthesis regulatory protein has a direct impact on the yield, production, and/or efficiency of production of lysine from that organism), or may have an indirect impact which nonetheless results in an increase in yield, production, and/or efficiency of production of the desired compound (e.g., where modulation of the regulation of a nucleotide biosynthesis protein has an impact on the production of an organic acid or a fatty acid from the bacterium, perhaps due to concomitant regulatory alterations in the biosynthetic or degradation pathways for these chemicals in response to the altered regulation of nucleotide biosynthesis). Aspects of the invention are further explicated below.
  • the term ‘fine chemical’ is art-recognized and includes molecules produced by an organism which have applications in various industries, such as, but not limited to, the pharmaceutical, agriculture, and cosmetics industries.
  • Such compounds include organic acids, such as tartaric acid, itaconic acid, and diaminopimelic acid, both proteinogenic and non-proteinogenic amino acids, purine and pyrimidine bases, nucleosides, and nucleotides (as described e.g. in Kuninaka, A. (1996) Nucleotides and related compounds, p. 561-612, in Biotechnology vol. 6, Rehm et al., eds.
  • VCH Weinheim, and references contained therein
  • lipids both saturated and unsaturated fatty acids (e.g., arachidonic acid), diols (e.g., propane diol, and butane diol), carbohydrates (e.g., hyaluronic acid and trehalose), aromatic compounds (e.g., aromatic amines, vanillin, and indigo), vitamins and cofactors (as described in Ullmann's Encyclopedia of Industrial Chemistry, vol. A27, “Vitamins”, p. 443-613 (1996) VCH: Weinheim and references therein; and Ong, A. S., Niki, E. & Packer, L.
  • Amino acids comprise the basic structural units of all proteins, and as such are essential for normal cellular functioning in all organisms.
  • the term “amino acid” is art-recognized.
  • the proteinogenic amino acids, of which there are 20 species, serve as structural units for proteins, in which they are linked by peptide bonds, while the nonproteinogenic amino acids (hundreds of which are known) are not normally found in proteins (see Ulmann's Encyclopedia of Industrial Chemistry, vol. A2, p. 57-97 VCH: Weinheim (1985)).
  • Amino acids may be in the D- or L-optical configuration, though L-amino acids are generally the only type found in naturally-occurring proteins.
  • the ‘essential’ amino acids histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, and valine
  • they are generally a nutritional requirement due to the complexity of their biosyntheses are readily converted by simple biosynthetic pathways to the remaining 11 ‘nonessential’ amino acids (alanine, arginine, asparagine, aspartate, cysteine, glutamate, glutamine, glycine, proline, serine, and tyrosine).
  • Higher animals do retain the ability to synthesize some of these amino acids, but the essential amino acids must be supplied from the diet in order for normal protein synthesis to occur.
  • Lysine is an important amino acid in the nutrition not only of humans, but also of monogastric animals such as poultry and swine.
  • Glutamate is most commonly used as a flavor additive (mono-sodium glutamate, MSG) and is widely used throughout the food industry, as are aspartate, phenylalanine, glycine, and cysteine. Glycine, L-methionine and tryptophan are all utilized in the pharmaceutical industry.
  • Glutamine, valine, leucine, isoleucine, histidine, arginine, proline, serine and alanine are of use in both the pharmaceutical and cosmetics industries. Threonine, tryptophan, and D/L-methionine are common feed additives. (Leuchtenberger, W. (1996) Amino aids—technical production and use, p. 466-502 in Rehm et al. (eds.) Biotechnology vol. 6, chapter 14a, VCH: Weinheim).
  • amino acids have been found to be useful as precursors for the synthesis of synthetic amino acids and proteins, such as N-acetylcysteine, S-carboxymethyl-L-cysteine, (S)-5-hydroxytryptophan, and others described in Ulmann's Encyclopedia of Industrial Chemistry, vol. A2, p. 57-97, VCH: Weinheim, 1985.
  • cysteine and glycine are produced from serine; the former by the condensation of homocysteine with serine, and the latter by the transferal of the side-chain ⁇ -carbon atom to tetrahydrofolate, in a reaction catalyzed by serine transhydroxymethylase.
  • Phenylalanine, and tyrosine are synthesized from the glycolytic and pentose phosphate pathway precursors erythrose 4-phosphate and phosphoenolpyruvate in a 9-step biosynthetic pathway that differ only at the final two steps after synthesis of prephenate. Tryptophan is also produced from these two initial molecules, but its synthesis is an 11-step pathway.
  • Tyrosine may also be synthesized from phenylalanine, in a reaction catalyzed by phenylalanine hydroxylase.
  • Alanine, valine, and leucine are all biosynthetic products of pyruvate, the final product of glycolysis.
  • Aspartate is formed from oxaloacetate, an intermediate of the citric acid cycle.
  • Asparagine, methionine, threonine, and lysine are each produced by the conversion of aspartate.
  • Isoleucine is formed from threonine.
  • a complex 9-step pathway results in the production of histidine from 5-phosphoribosyl-1-pyrophosphate, an activated sugar.
  • Amino acids in excess of the protein synthesis needs of the cell cannot be stored, and are instead degraded to provide intermediates for the major metabolic pathways of the cell (for review see Stryer, L. Biochemistry 3 rd ed. Ch. 21 “Amino Acid Degradation and the Urea Cycle” p. 495-516 (1988)).
  • the cell is able to convert unwanted amino acids into useful metabolic intermediates, amino acid production is costly in terms of energy, precursor molecules, and the enzymes necessary to synthesize them.
  • amino acid biosynthesis is regulated by feedback inhibition, in which the presence of a particular amino acid serves to slow or entirely stop its own production (for overview of feedback mechanisms in amino acid biosynthetic pathways, see Stryer, L. Biochemistry, 3 rd ed. Ch. 24: “Biosynthesis of Amino Acids and Heme” p. 575-600 (1988)).
  • the output of any particular amino acid is limited by the amount of that amino acid present in the cell.
  • Vitamins, cofactors, and nutraceuticals comprise another group of molecules which the higher animals have lost the ability to synthesize and so must ingest, although they are readily synthesized by other organisms such as bacteria. These molecules are either bioactive substances themselves, or are precursors of biologically active substances which may serve as electron carriers or intermediates in a variety of metabolic pathways. Aside from their nutritive value, these compounds also have significant industrial value as coloring agents, antioxidants, and catalysts or other processing aids. (For an overview of the structure, activity, and industrial applications of these compounds, see, for example, Ullman's Encyclopedia of Industrial Chemistry, “Vitamins” vol. A27, p.
  • vitamin is art-recognized, and includes nutrients which are required by an organism for normal functioning, but which that organism cannot synthesize by itself.
  • the group of vitamins may encompass cofactors and nutraceutical compounds.
  • cofactor includes nonproteinaceous compounds required for a normal enzymatic activity to occur. Such compounds may be organic or inorganic; the cofactor molecules of the invention are preferably organic.
  • nutraceutical includes dietary supplements having health benefits in plants and animals, particularly humans. Examples of such molecules are vitamins, antioxidants, and also certain lipids (e.g., polyunsaturated fatty acids).
  • Thiamin (vitamin B 1 ) is produced by the chemical coupling of pyrimidine and thiazole moieties.
  • Riboflavin (vitamin B 2 ) is synthesized from guanosine-5′-triphosphate (GTP) and ribose-5′-phosphate. Riboflavin, in turn, is utilized for the synthesis of flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD).
  • vitamin B 6 The family of compounds collectively termed ‘vitamin B 6 ’ (e.g., pyridoxine, pyridoxamine, pyridoxa-5′-phosphate, and the commercially used pyridoxin hydrochloride) are all derivatives of the common structural unit, 5-hydroxy-6-methylpyridine.
  • Pantothenate pantothenic acid, (R)-(+)-N-(2,4-dihydroxy-3,3-dimethyl-1-oxobutyl)- ⁇ -alanine
  • pantothenate biosynthesis consist of the ATP-driven condensation of ⁇ -alanine and pantoic acid.
  • pantothenate The enzymes responsible for the biosynthesis steps for the conversion to pantoic acid, to ⁇ -alanine and for the condensation to panthotenic acid are known.
  • the metabolically active form of pantothenate is Coenzyme A, for which the biosynthesis proceeds in 5 enzymatic steps.
  • Pantothenate, pyridoxal-5′-phosphate, cysteine and ATP are the precursors of Coenzyme A.
  • These enzymes not only catalyze the formation of panthothante, but also the production of (R)-pantoic acid, (R)-pantolacton, (R)-panthenol (provitamin B 5 ), pantetheine (and its derivatives) and coenzyme A.
  • Biotin biosynthesis from the precursor molecule pimeloyl-CoA in microorganisms has been studied in detail and several of the genes involved have been identified. Many of the corresponding proteins have been found to also be involved in Fe-cluster synthesis and are members of the nifS class of proteins.
  • Lipoic acid is derived from octanoic acid, and serves as a coenzyme in energy metabolism, where it becomes part of the pyruvate dehydrogenase complex and the ⁇ -ketoglutarate dehydrogenase complex.
  • the folates are a group of substances which are all derivatives of folic acid, which is turn is derived from L-glutamic acid, p-amino-benzoic acid and 6-methylpterin.
  • GTP guanosine-5′-triphosphate
  • L-glutamic acid L-glutamic acid
  • p-amino-benzoic acid The biosynthesis of folic acid and its derivatives, starting from the metabolism intermediates guanosine-5′-triphosphate (GTP), L-glutamic acid and p-amino-benzoic acid has been studied in detail in certain microorganisms.
  • Corrinoids such as the cobalamines and particularly vitamin B 12
  • porphyrines belong to a group of chemicals characterized by a tetrapyrole ring system.
  • the biosynthesis of vitamin B 12 is sufficiently complex that it has not yet been completely characterized, but many of the enzymes and substrates involved are now known.
  • Nicotinic acid (nicotinate), and nicotinamide are pyridine derivatives which are also termed ‘niacin’.
  • Niacin is the precursor of the important coenzymes NAD (nicotinamide adenine dinucleotide) and NADP (nicotinamide adenine dinucleotide phosphate) and their reduced forms.
  • purine and pyrimidine metabolism genes and their corresponding proteins are important targets for the therapy of tumor diseases and viral infections.
  • the language “purine” or “pyrimidine” includes the nitrogenous bases which are constituents of nucleic acids, co-enzymes, and nucleotides.
  • the term “nucleotide” includes the basic structural units of nucleic acid molecules, which are comprised of a nitrogenous base, a pentose sugar (in the case of RNA, the sugar is ribose; in the case of DNA, the sugar is D-deoxyribose), and phosphoric acid.
  • the language “nucleoside” includes molecules which serve as precursors to nucleotides, but which are lacking the phosphoric acid moiety that nucleotides possess.
  • nucleic acid molecules By inhibiting the biosynthesis of these molecules, or their mobilization to form nucleic acid molecules, it is possible to inhibit RNA and DNA synthesis; by inhibiting this activity in a fashion targeted to cancerous cells, the ability of tumor cells to divide and replicate may be inhibited. Additionally, there are nucleotides which do not form nucleic acid molecules, but rather serve as energy stores (i.e., AMP) or as coenzymes (i.e., FAD and NAD).
  • energy stores i.e., AMP
  • coenzymes i.e., FAD and NAD
  • purine and pyrimidine bases, nucleosides and nucleotides have other utilities: as intermediates in the biosynthesis of several fine chemicals (e.g., thiamine, S-adenosyl-methionine, folates, or riboflavin), as energy carriers for the cell (e.g., ATP or GTP), and for chemicals themselves, commonly used as flavor enhancers (e.g., IMP or GMP) or for several medicinal applications (see, for example, Kuninaka, A. (1996) Nucleotides and Related Compounds in Biotechnology vol. 6, Rehm et al., eds. VCH: Weinheim, p. 561-612).
  • enzymes involved in purine, pyrimidine, nucleoside, or nucleotide metabolism are increasingly serving as targets against which chemicals for crop protection, including fungicides, herbicides and insecticides, are developed.
  • Purine nucleotides are synthesized from ribose-5-phosphate, in a series of steps through the intermediate compound inosine-5′-phosphate (IMP), resulting in the production of guanosine-5′-monophosphate (GMP) or adenosine-5′-monophosphate (AMP), from which the triphosphate forms utilized as nucleotides are readily formed. These compounds are also utilized as energy stores, so their degradation provides energy for many different biochemical processes in the cell. Pyrimidine biosynthesis proceeds by the formation of uridine-5′-monophosphate (UMP) from ribose-5-phosphate. UMP, in turn, is converted to cytidine-5′-triphosphate (CTP).
  • IMP inosine-5′-phosphate
  • AMP adenosine-5′-monophosphate
  • the deoxy-forms of all of these nucleotides are produced in a one step reduction reaction from the diphosphate ribose form of the nucleotide to the diphosphate deoxyribose form of the nucleotide. Upon phosphorylation, these molecules are able to participate in DNA synthesis.
  • Trehalose consists of two glucose molecules, bound in ⁇ , ⁇ -1,1 linkage. It is commonly used in the food industry as a sweetener, an additive for dried or frozen foods, and in beverages. However, it also has applications in the pharmaceutical, cosmetics and biotechnology industries (see, for example, Nishimoto et al., (1998) U.S. Pat. No. 5,759,610; Singer, M. A. and Lindquist, S. (1998) Trends Biotech. 16: 460-467; Paiva, C. L. A. and Panek, A. D. (1996) Biotech. Ann. Rev. 2: 293-314; and Shiosaka, M. (1997) J. Japan 172: 97-102). Trehalose is produced by enzymes from many microorganisms and is naturally released into the surrounding medium, from which it can be collected using methods known in the art.
  • All living cells have complex catabolic and anabolic metabolic capabilities with many interconnected pathways.
  • the cell employs a finely-tuned regulatory network. By regulating enzyme synthesis and enzyme activity, either independently or simultaneously, the cell is able to control the activity of disparate metabolic pathways to reflect the changing needs of the cell.
  • the induction or repression of enzyme synthesis may occur at either the level of transcription or translation, or both.
  • Gene expression in prokaryotes is regulated by several mechanisms at the level of transcription (for review see e.g., Lewin, B (1990) Genes IV, Part 3: “Controlling prokaryotic genes by transcription”, Oxford University Press: Oxford, p. 213-301, and references therein, and Michal, G. (1999) Biochemical Pathways: An Atlas of Biochemistry and Molecular Biology, John Wiley & Sons). All such known regulatory processes are mediated by additional genes, which themselves respond to external influences of various kinds (e.g., temperature, nutrient availability, or light). Exemplary protein factors which have been implicated in this type of regulation include the transcription factors.
  • proteins which bind to DNA thereby either increasing the expression of a gene (positive regulation, as in the case of e.g. the ara operon from E. coli ) or decreasing gene expression (negative regulation, as in the case of the lac operon from E. coli ).
  • These expression-modulating transcription factors can themselves be the subject of regulation. Their activity can, for example, be regulated by the binding of low molecular weight compounds to the DNA-binding protein, thereby stimulating (as in the case of arabinose for the ara operon) or inhibiting (as in the case of the lactose for the lac operon) the binding of these proteins to the appropriate binding site on the DNA (see, for example, Helmann, J. D. and Chamberlin, M. J.
  • protein synthesis is also often regulated at the level of translation.
  • mechanisms by which such regulation may occur including alteration of the ability of the ribosome to bind to one or more mRNAs, binding of the ribosome to the mRNA, the maintenance or removal of mRNA secondary structure, the utilization of common or less common codons for a particular gene, the degree of abundance of one or more tRNAs, and special regulation mechanisms, such as attenuation (see Vellanoweth, R. I. (1993) Translation and its regulation in Bacillus subtilis and other gram-positive bacteria, Sonenshein, A. L. et al., eds. ASM: Washington, D.C., p. 699-711 and references cited therein).
  • Transcriptional and translational regulation may be targeted to a single protein (sequential regulation) or simultaneously to several proteins in different metabolic pathways (coordinate regulation). Often, genes whose expression is coordinately regulated are physically located near one another in the genome, in an operon or regulon. Such up- or down-regulation of gene transcription and translation is governed by the cellular and extracellular levels of various factors, such as substrates (precursor and intermediate molecules used in one or more metabolic pathways), catabolites (molecules produced by biochemical pathways concerned with the production of energy from the breakdown of complex organic molecules such as sugars), and end products (the molecules resulting at the end of a metabolic pathway).
  • substrates precursor and intermediate molecules used in one or more metabolic pathways
  • catabolites molecules produced by biochemical pathways concerned with the production of energy from the breakdown of complex organic molecules such as sugars
  • end products the molecules resulting at the end of a metabolic pathway.
  • genes encoding enzymes necessary for the activity of a particular pathway is induced by high levels of substrate molecules for that pathway. Similarly, such gene expression tends to be repressed when there exist high intracellular levels of the end product of the pathway (Snyder, L. and Champness, W. (1997) The Molecular Biology of Bacteria ASM: Washington).
  • Gene expression may also be regulated by other external and internal factors, such as environmental conditions (e.g., heat, oxidative stress, or starvation). These global environmental changes cause alterations in the expression of specialized modulating genes, which directly or indirectly (via additional genes or proteins) trigger the expression of genes by means of binding to DNA and thereby inducing or repressing transcription (see, for example, Lin, E. C. C. and Lynch, A. S., eds. (1995) Regulation of Gene Expression in Escherichia coli . Chapman & Hall: New York).
  • cellular metabolism may be regulated.
  • Such regulation is accomplished either by the activities of other proteins, or by binding of low-molecular-weight components which either impede or enable the normal functioning of the protein.
  • Examples of protein regulation by the binding of low-molecular-weight compounds include the binding of GTP or NAD.
  • the binding of a low-molecular-weight chemical is typically reversible, as is the case with the GTP-binding proteins. These proteins exist in two stages (with bound GTP or GDP), one stage being the activated form of the protein, and one stage being inactive.
  • Regulation of protein activity by the action of other enzymes typically takes the form of covalent modification of the protein (i.e., phosphorylation of amino acid residues such as histidine or aspartate, or methylation).
  • covalent modification is typically reversible, as mediated by an enzyme of the opposite activity.
  • An example of this is the opposite activities of kinases and phosphorylases in protein phosphorylation; protein kinases phosphorylate specific residues on a target protein (e.g., serine or threonine), while protein phosphorylases remove phosphate groups from such proteins.
  • enzymes which modulate the activity of other proteins are themselves modulated by external stimuli. These stimuli are mediated through proteins which function as sensors.
  • the present invention is based, at least in part, on the discovery of novel molecules, referred to herein as MR nucleic acid and protein molecules, which regulate, by transcriptional, translational, or post-translational means, one or more metabolic pathways in C. glutamicum .
  • the MR molecules transcriptionally, translationally, or posttranslationally regulate a metabolic pathway in C. glutamicum .
  • the activity of the MR molecules of the present invention to regulate one or more C. glutamicum metabolic pathways has an impact on the production of a desired fine chemical by this organism.
  • the MR molecules of the invention are modulated in activity, such that the C. glutamicum metabolic pathways which the MR proteins of the invention regulate are modulated in efficiency or output, which either directly or indirectly modulates the yield, production, and/or efficiency of production of a desired fine chemical by C. glutamicum.
  • MR protein or “MR polypeptide” includes proteins which transcriptionally, translationally, or posttranslationally regulate a metabolic pathway in C. glutamicum .
  • MR proteins include those encoded by the MR genes set forth in Table 1 and Appendix A.
  • MR gene or “MR nucleic acid sequence” include nucleic acid sequences encoding an MR protein, which consist of a coding region and also corresponding untranslated 5′ and 3′ sequence regions. Examples of MR genes include those set forth in Table 1.
  • production or “productivity” are art-recognized and include the concentration of the fermentation product (for example, the desired fine chemical) formed within a given time and a given fermentation volume (e.g., kg product per hour per liter).
  • the term “efficiency of production” includes the time required for a particular level of production to be achieved (for example, how long it takes for the cell to attain a particular rate of output of a fine chemical).
  • yield or “product/carbon yield” is art-recognized and includes the efficiency of the conversion of the carbon source into the product (i.e., fine chemical). This is generally written as, for example, kg product per kg carbon source.
  • biosynthesis or a “biosynthetic pathway” are art-recognized and include the synthesis of a compound, preferably an organic compound, by a cell from intermediate compounds in what may be a multistep and highly regulated process.
  • degradation or a “degradation pathway” are art-recognized and include the breakdown of a compound, preferably an organic compound, by a cell to degradation products (generally speaking, smaller or less complex molecules) in what may be a multistep and highly regulated process.
  • the language “metabolism” is art-recognized and includes the totality of the biochemical reactions that take place in an organism.
  • the metabolism of a particular compound comprises the overall biosynthetic, modification, and degradation pathways in the cell related to this compound.
  • regulation is art-recognized and includes the activity of a protein to govern the activity of another protein.
  • transcriptional regulation is art-recognized and includes the activity of a protein to impede or activate the conversion of a DNA encoding a target protein to mRNA.
  • translational regulation is art-recognized and includes the activity of a protein to impede or activate the conversion of an mRNA encoding a target protein to a protein molecule.
  • posttranslational regulation includes the activity of a protein to impede or improve the activity of a target protein by covalently modifying the target protein (e.g., by methylation, glucosylation, or phosphorylation).
  • the MR molecules of the invention are capable of modulating the production of a desired molecule, such as a fine chemical, in a microorganism such as C. glutamicum .
  • a desired molecule such as a fine chemical
  • a microorganism such as C. glutamicum
  • one or more of the regulatory proteins of the invention for metabolic pathways may be manipulated such that its function is modulated.
  • a biosynthetic enzyme may be improved in efficiency, or its allosteric control region destroyed such that feedback inhibition of production of the compound is prevented.
  • a degradative enzyme may be deleted or modified by substitution, deletion, or addition such that its degradative activity is lessened for the desired compound without impairing the viability of the cell. In each case, the overall yield or rate of production of one of these desired fine chemicals may be increased.
  • MR proteins of the invention may uncouple the growth and division of cells from their extracellular surroundings to a certain degree; by impairing an MR protein which normally represses biosynthesis of a nucleotide when the extracellular conditions are suboptimal for growth and cell division such that it now lacks this function, one may permit growth to occur even when the extracellular conditions are poor.
  • This is of particular relevance in large-scale fermentative growth, where conditions within the culture are often suboptimal in terms of temperature, nutrient supply or aeration, but would still support growth and cell division if the cellular regulatory systems for these factors were eliminated.
  • the isolated nucleic acid sequences of the invention are contained within the genome of a Corynebacterium glutamicum strain available through the American Type Culture Collection, given designation ATCC 13032.
  • the nucleotide sequence of the isolated C. glutamicum MR DNAs and the predicted amino acid sequences of the C. glutamicum MR proteins are shown in Appendices A and B, respectively. Computational analyses were performed which classified and/or identified these nucleotide sequences as sequences which encode metabolic pathway regulatory proteins.
  • the present invention also pertains to proteins which have an amino acid sequence which is substantially homologous to an amino acid sequence of Appendix B.
  • a protein which has an amino acid sequence which is substantially homologous to a selected amino acid sequence is least about 50% homologous to the selected amino acid sequence, e.g., the entire selected amino acid sequence.
  • a protein which has an amino acid sequence which is substantially homologous to a selected amino acid sequence can also be least about 50-60%, preferably at least about 60-70%. and more preferably at least about 70-80%, 80-90%, or 90-95%, and most preferably at least about 96%, 97%, 98%, 99% or more homologous to the selected amino acid sequence.
  • the MR protein or a biologically active portion or fragment thereof of the invention can transcriptionally, translationally, or posttranslationally regulate a metabolic pathway in C. glutamicum , or have one or more of the activities set forth in Table 1.
  • nucleic acid molecules that encode MR polypeptides or biologically active portions thereof, as well as nucleic acid fragments sufficient for use as hybridization probes or primers for the identification or amplification of MR-encoding nucleic acid (e.g., MR DNA).
  • nucleic acid molecule is intended to include DNA molecules (e.g., cDNA or genomic DNA) and RNA molecules (e.g., mRNA) and analogs of the DNA or RNA generated using nucleotide analogs.
  • nucleic acid molecule can be single-stranded or double-stranded, but preferably is double-stranded DNA.
  • isolated nucleic acid molecule is one which is separated from other nucleic acid molecules which are present in the natural source of the nucleic acid.
  • an “isolated” nucleic acid is free of sequences which naturally flank the nucleic acid (i.e., sequences located at the 5′ and 3′ ends of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived.
  • the isolated MR nucleic acid molecule can contain less than about 5 kb, 4 kb, 3 kb, 2 kb, 1 kb, 0.5 kb or 0.1 kb of nucleotide sequences which naturally flank the nucleic acid molecule in genomic DNA of the cell from which the nucleic acid is derived (e.g., a C. glutamicum cell).
  • an “isolated” nucleic acid molecule, such as a DNA molecule can be substantially free of other cellular material, or culture medium when produced by recombinant techniques, or chemical precursors or other chemicals when chemically synthesized.
  • a nucleic acid molecule of the present invention e.g., a nucleic acid molecule having a nucleotide sequence of Appendix A, or a portion thereof, can be isolated using standard molecular biology techniques and the sequence information provided herein.
  • a C. glutamicum MR DNA can be isolated from a C. glutamicum library using all or portion of one of the sequences of Appendix A as a hybridization probe and standard hybridization techniques (e.g., as described in Sambrook, J., Fritsh, E. F., and Maniatis, T. Molecular Cloning: A Laboratory Manual. 2 nd, ed ., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989).
  • nucleic acid molecule encompassing all or a portion of one of the sequences of Appendix A can be isolated by the polymerase chain reaction using oligonucleotide primers designed based upon this sequence (e.g., a nucleic acid molecule encompassing all or a portion of one of the sequences of Appendix A can be isolated by the polymerase chain reaction using oligonucleotide primers designed based upon this same sequence of Appendix A).
  • mRNA can be isolated from normal endothelial cells (e.g., by the guanidinium-thiocyanate extraction procedure of Chirgwin et al.
  • DNA can be prepared using reverse transcriptase (e.g., Moloney MLV reverse transcriptase, available from Gibco/BRL, Bethesda, Md.; or AMV reverse transcriptase, available from Seikagaku America, Inc., St. Louis, Fla.).
  • reverse transcriptase e.g., Moloney MLV reverse transcriptase, available from Gibco/BRL, Bethesda, Md.; or AMV reverse transcriptase, available from Seikagaku America, Inc., St. Russia, Fla.
  • Synthetic oligonucleotide primers for polymerase chain reaction amplification can be designed based upon one of the nucleotide sequences shown in Appendix A.
  • a nucleic acid of the invention can be amplified using cDNA or, alternatively, genomic DNA, as a template and appropriate oligonucleotide primers according to standard PCR amplification techniques.
  • nucleic acid so amplified can be cloned into an appropriate vector and characterized by DNA sequence analysis.
  • oligonucleotides corresponding to an MR nucleotide sequence can be prepared by standard synthetic techniques, e.g., using an automated DNA synthesizer.
  • an isolated nucleic acid molecule of the invention comprises one of the nucleotide sequences shown in Appendix A.
  • the sequences of Appendix A correspond to the Corynebacterium glutamicum MR DNAs of the invention.
  • This DNA comprises sequences encoding MR proteins (i.e., the “coding region”, indicated in each sequence in Appendix A), as well as 5′ untranslated sequences and 3′ untranslated sequences, also indicated in Appendix A.
  • the nucleic acid molecule can comprise only the coding region of any of the sequences in Appendix A.
  • each of the sequences set forth in Appendix A has an identifying RXA, RXN, or RXS number having the designation “RXA”, “RXN”, or “RXS” followed by 5 digits (i.e., RXA00603, RXN03181, or RXS00686).
  • Each of these sequences comprises up to three parts: a 5′ upstream region, a coding region, and a downstream region. Each of these three regions is identified by the same RXA, RXN, or RXS designation to eliminate confusion.
  • the amino acid sequences in Appendix B designated RXA00603, RXN03181, and RXS00686 are translations of the coding regions of the nucleotide sequence of nucleic acid molecules RXA00603, RXN03181, and RXS00686, respectively, in Appendix A.
  • Each of the RXA, RXN, and RXS nucleotide and amino acid sequences of the invention has also been assigned a SEQ ID NO, as indicated in Table 1.
  • Table 1 the nucleotide sequence of RXA00603 is SEQ ID NO:5 and the amino acid sequence of RXA00603 is SEQ ID NO: 6.
  • F-designated genes include those genes set forth in Table 1 which have an ‘F’ in front of the RXA, RXN, or RXS designation.
  • SEQ ID NO:3 designated, as indicated on Table 1, as “F RXA02880”, is an F-designated gene, as are SEQ ID NOs: 21, 27, and 33 (designated on Table 1 as “F RXA02493”, “F RXA00291”, and “F RXA00651”, respectively).
  • the nucleic acid molecules of the present invention are not intended to include those compiled in Table 2.
  • a sequence for this gene was published in Wehrmann, A., et al. (1998) J. Bacteriol. 180(12): 3159-3165.
  • the sequence obtained by the inventors of the present application is significantly longer than the published version. It is believed that the published version relied on an incorrect start codon, and thus represents only a fragment of the actual coding region.
  • an isolated nucleic acid molecule of the invention comprises a nucleic acid molecule which is a complement of one of the nucleotide sequences shown in Appendix A, or a portion thereof.
  • a nucleic acid molecule which is complementary to one of the nucleotide sequences shown in Appendix A is one which is sufficiently complementary to one of the nucleotide sequences shown in Appendix A such that it can hybridize to one of the nucleotide sequences shown in Appendix A, thereby forming a stable duplex.
  • an isolated nucleic acid molecule of the invention comprises a nucleotide sequence which is at least about 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, or 60%, preferably at least about 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, or 70%, more preferably at least about 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, or 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, or 90%, or 91%, 92%, 93%, 94%, and even more preferably at least about 95%, 96%, 97%, 98%, 99% or more homologous to a nucleotide sequence shown in Appendix A, or a portion thereof.
  • an isolated nucleic acid molecule of the invention comprises a nucleotide sequence which hybridizes, e.g., hybridizes under stringent conditions, to one of the nucleotide sequences shown in Appendix A, or a portion thereof.
  • the nucleic acid molecule of the invention can comprise only a portion of the coding region of one of the sequences in Appendix A, for example a fragment which can be used as a probe or primer or a fragment encoding a biologically active portion of an MR protein.
  • the nucleotide sequences determined from the cloning of the MR genes from C. glutamicum allows for the generation of probes and primers designed for use in identifying and/or cloning MR homologues in other cell types and organisms, as well as MR homologues from other Corynebacteria or related species.
  • the probe/primer typically comprises substantially purified oligonucleotide.
  • the oligonucleotide typically comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least about 12, preferably about 25, more preferably about 40, 50 or 75 consecutive nucleotides of a sense strand of one of the sequences set forth in Appendix A, an anti-sense sequence of one of the sequences set forth in Appendix A, or naturally occurring mutants thereof.
  • Primers based on a nucleotide sequence of Appendix A can be used in PCR reactions to clone MR homologues. Probes based on the MR nucleotide sequences can be used to detect transcripts or genomic sequences encoding the same or homologous proteins.
  • the probe further comprises a label group attached thereto, e.g. the label group can be a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor.
  • the label group can be a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor.
  • Such probes can be used as a part of a diagnostic test kit for identifying cells which misexpress an MR protein, such as by measuring a level of an MR-encoding nucleic acid in a sample of cells, e.g., detecting MR mRNA levels or determining whether a genomic MR gene has been mutated or deleted.
  • the nucleic acid molecule of the invention encodes a protein or portion thereof which includes an amino acid sequence which is sufficiently homologous to an amino acid sequence of Appendix B such that the protein or portion thereof maintains the ability to transcriptionally, translationally, or posttranslationally regulate a metabolic pathway in C. glutamicum .
  • the language “sufficiently homologous” refers to proteins or portions thereof which have amino acid sequences which include a minimum number of identical or equivalent (e.g., an amino acid residue which has a similar side chain as an amino acid residue in one of the sequences of Appendix B) amino acid residues to an amino acid sequence of Appendix B such that the protein or portion thereof is able to transcriptionally, translationally, or posttranslationally regulate a metabolic pathway in C. glutamicum . Protein members of such metabolic pathways, as described herein, may function to regulate the biosynthesis or degradation of one or more fine chemicals. Examples of such activities are also described herein.
  • MR protein contributes to the overall regulation of one or more fine chemical metabolic pathway, or contributes, either directly or indirectly, to the yield, production, and/or efficiency of production of one or more fine chemicals.
  • Examples of MR protein activities are set forth in Table 1.
  • the protein is at least about 50-60%, preferably at least about 60-70%, and more preferably at least about 70-80%, 80-90%, 90-95%, and most preferably at least about 96%, 97%, 98%, 99% or more homologous to an entire amino acid sequence of Appendix B.
  • portions of proteins encoded by the MR nucleic acid molecules of the invention are preferably biologically active portions of one of the MR proteins.
  • biologically active portion of an MR protein is intended to include a portion, e.g., a domain/motif, of an MR protein that transcriptionally, translationally, or posttranslationally regulates a metabolic pathway in C. glutamicum , or has an activity as set forth in Table 1.
  • an assay of enzymatic activity may be performed. Such assay methods are well known to those of ordinary skill in the art, as detailed in Example 8 of the Exemplification.
  • Additional nucleic acid fragments encoding biologically active portions of an MR protein can be prepared by isolating a portion of one of the sequences in Appendix B, expressing the encoded portion of the MR protein or peptide (e.g., by recombinant expression in vitro) and assessing the activity of the encoded portion of the MR protein or peptide.
  • the invention further encompasses nucleic acid molecules that differ from one of the nucleotide sequences shown in Appendix A (and portions thereof) due to degeneracy of the genetic code and thus encode the same MR protein as that encoded by the nucleotide sequences shown in Appendix A.
  • an isolated nucleic acid molecule of the invention has a nucleotide sequence encoding a protein having an amino acid sequence shown in Appendix B.
  • the nucleic acid molecule of the invention encodes a full length C. glutamicum protein which is substantially homologous to an amino acid sequence of Appendix B (encoded by an open reading frame shown in Appendix A).
  • sequences of the invention are not meant to include the sequences of the prior art, such as those Genbank sequences set forth in Tables 2 or 4 which were available prior to the present invention.
  • the invention includes nucleotide and amino acid sequences having a percent identity to a nucleotide or amino acid sequence of the invention which is greater than that of a sequence of the prior art (e.g., a Genbank sequence (or the protein encoded by such a sequence) set forth in Tables 2 or 4).
  • the invention includes a nucleotide sequence which is greater than and/or at least 40% identical to the nucleotide sequence designated RXA00603 (SEQ ID NO:5), a nucleotide sequence which is greater than and/or at least 55% identical to the nucleotide sequence designated RXA00129 (SEQ ID NO:29), and a nucleotide sequence which is greater than and/or at least 40% identical to the nucleotide sequence designated RXA00006 (SEQ ID NO:35).
  • nucleic acid and amino acid sequences having percent identities greater than the lower threshold so calculated are also encompassed by the invention.
  • DNA sequence polymorphisms that lead to changes in the amino acid sequences of MR proteins may exist within a population (e.g., the C. glutamicum population).
  • Such genetic polymorphism in the MR gene may exist among individuals within a population due to natural variation.
  • the terms “gene” and “recombinant gene” refer to nucleic acid molecules comprising an open reading frame encoding an MR protein, preferably a C. glutamicum MR protein.
  • Such natural variations can typically result in 1-5% variance in the nucleotide sequence of the MR gene. Any and all such nucleotide variations and resulting amino acid polymorphisms in MR that are the result of natural variation and that do not alter the functional activity of MR proteins are intended to be within the scope of the invention.
  • Nucleic acid molecules corresponding to natural variants and non- C. glutamicum homologues of the C. glutamicum MR DNA of the invention can be isolated based on their homology to the C. glutamicum MR nucleic acid disclosed herein using the C. glutamicum DNA, or a portion thereof, as a hybridization probe according to standard hybridization techniques under stringent hybridization conditions. Accordingly, in another embodiment, an isolated nucleic acid molecule of the invention is at least 15 nucleotides in length and hybridizes under stringent conditions to the nucleic acid molecule comprising a nucleotide sequence of Appendix A. In other embodiments, the nucleic acid is at least 30, 50, 100, 250 or more nucleotides in length.
  • hybridizes under stringent conditions is intended to describe conditions for hybridization and washing under which nucleotide sequences at least 60% homologous to each other typically remain hybridized to each other.
  • the conditions are such that sequences at least about 65%, more preferably at least about 70%, and even more preferably at least about 75% or more homologous to each other typically remain hybridized to each other.
  • stringent conditions are known to those of ordinary skill in the art and can be found in Current Protocols in Molecular Biology , John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6.
  • a preferred, non-limiting example of stringent hybridization conditions are hybridization in 6 ⁇ sodium chloride/sodium citrate (SSC) at about 45° C., followed by one or more washes in 0.2 ⁇ SSC, 0.1% SDS at 50-65° C.
  • an isolated nucleic acid molecule of the invention that hybridizes under stringent conditions to a sequence of Appendix A corresponds to a naturally-occurring nucleic acid molecule.
  • a “naturally-occurring” nucleic acid molecule refers to an RNA or DNA molecule having a nucleotide sequence that occurs in nature (e.g., encodes a natural protein).
  • the nucleic acid encodes a natural C. glutamicum MR protein.
  • nucleotide sequence of Appendix A thereby leading to changes in the amino acid sequence of the encoded MR protein, without altering the functional ability of the MR protein.
  • nucleotide substitutions leading to amino acid substitutions at “non-essential” amino acid residues can be made in a sequence of Appendix A.
  • a “non-essential” amino acid residue is a residue that can be altered from the wild-type sequence of one of the MR proteins (Appendix B) without altering the activity of said MR protein, whereas an “essential” amino acid residue is required for MR protein activity.
  • Other amino acid residues, however, may not be essential for activity and thus are likely to be amenable to alteration without altering MR activity.
  • nucleic acid molecules encoding MR proteins that contain changes in amino acid residues that are not essential for MR activity. Such MR proteins differ in amino acid sequence from a sequence contained in Appendix B yet retain at least one of the MR activities described herein.
  • the isolated nucleic acid molecule comprises a nucleotide sequence encoding a protein, wherein the protein comprises an amino acid sequence at least about 50% homologous to an amino acid sequence of Appendix B and is capable of transcriptionally, translationally, or posttranslationally regulating a metabolic pathway in C. glutamicum , or has one or more activities set forth in Table 1.
  • the protein encoded by the nucleic acid molecule is at least about 50-60% homologous to one of the sequences in Appendix B, more preferably at least about 60-70% homologous to one of the sequences in Appendix B, even more preferably at least about 70-80%, 80-90%, 90-95% homologous to one of the sequences in Appendix B, and most preferably at least about 96%, 97%, 98%, or 99% homologous to one of the sequences in Appendix B.
  • sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of one protein or nucleic acid for optimal alignment with the other protein or nucleic acid).
  • amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared.
  • a position in one sequence e.g., one of the sequences of Appendix B
  • the molecules are homologous at that position (i.e., as used herein amino acid or nucleic acid “homology” is equivalent to amino acid or nucleic acid “identity”).
  • An isolated nucleic acid molecule encoding an MR protein homologous to a protein sequence of Appendix B can be created by introducing one or more nucleotide substitutions, additions or deletions into a nucleotide sequence of Appendix A such that one or more amino acid substitutions, additions or deletions are introduced into the encoded protein. Mutations can be introduced into one of the sequences of Appendix A by standard techniques, such as site-directed mutagenesis and PCR-mediated mutagenesis. Preferably, conservative amino acid substitutions are made at one or more predicted non-essential amino acid residues. A “conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain.
  • Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine).
  • basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.g.
  • a predicted nonessential amino acid residue in an MR protein is preferably replaced with another amino acid residue from the same side chain family.
  • mutations can be introduced randomly along all or part of an MR coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for an MR activity described herein to identify mutants that retain MR activity.
  • the encoded protein can be expressed recombinantly and the activity of the protein can be determined using, for example, assays described herein (see Example 8 of the Exemplification).
  • an antisense nucleic acid comprises a nucleotide sequence which is complementary to a “sense” nucleic acid encoding a protein, e.g., complementary to the coding strand of a double-stranded DNA molecule or complementary to an mRNA sequence. Accordingly, an antisense nucleic acid can hydrogen bond to a sense nucleic acid.
  • the antisense nucleic acid can be complementary to an entire MR coding strand, or to only a portion thereof.
  • an antisense nucleic acid molecule is antisense to a “coding region” of the coding strand of a nucleotide sequence encoding an MR protein.
  • coding region refers to the region of the nucleotide sequence comprising codons which are translated into amino acid residues (e.g., the entire coding region of SEQ ID NO: 1 (RXN03181) comprises nucleotides 1 to 414).
  • the antisense nucleic acid molecule is antisense to a “noncoding region” of the coding strand of a nucleotide sequence encoding MR.
  • noncoding region refers to 5′ and 3′ sequences which flank the coding region that are not translated into amino acids (i.e., also referred to as 5′ and 3′ untranslated regions).
  • antisense nucleic acids of the invention can be designed according to the rules of Watson and Crick base pairing.
  • the antisense nucleic acid molecule can be complementary to the entire coding region of MR mRNA, but more preferably is an oligonucleotide which is antisense to only a portion of the coding or noncoding region of MR mRNA.
  • the antisense oligonucleotide can be complementary to the region surrounding the translation start site of MR mRNA.
  • An antisense oligonucleotide can be, for example, about 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 nucleotides in length.
  • An antisense nucleic acid of the invention can be constructed using chemical synthesis and enzymatic ligation reactions using procedures known in the art.
  • an antisense nucleic acid e.g., an antisense oligonucleotide
  • an antisense nucleic acid e.g., an antisense oligonucleotide
  • modified nucleotides which can be used to generate the antisense nucleic acid include 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl)uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D-mannosylqueosine, 5′-methoxycarbox
  • the antisense nucleic acid can be produced biologically using an expression vector into which a nucleic acid has been subcloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest, described further in the following subsection).
  • the antisense nucleic acid molecules of the invention are typically administered to a cell or generated in situ such that they hybridize with or bind to cellular mRNA and/or genomic DNA encoding an MR protein to thereby inhibit expression of the protein, e.g., by inhibiting transcription and/or translation.
  • the hybridization can be by conventional nucleotide complementarity to form a stable duplex, or, for example, in the case of an antisense nucleic acid molecule which binds to DNA duplexes, through specific interactions in the major groove of the double helix.
  • the antisense molecule can be modified such that it specifically binds to a receptor or an antigen expressed on a selected cell surface, e.g., by linking the antisense nucleic acid molecule to a peptide or an antibody which binds to a cell surface receptor or antigen.
  • the antisense nucleic acid molecule can also be delivered to cells using the vectors described herein. To achieve sufficient intracellular concentrations of the antisense molecules, vector constructs in which the antisense nucleic acid molecule is placed under the control of a strong prokaryotic, viral, or eukaryotic promoter are preferred.
  • the antisense nucleic acid molecule of the invention is an ⁇ -anomeric nucleic acid molecule.
  • An ⁇ -anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual ⁇ -units, the strands run parallel to each other (Gaultier et al. (1987) Nucleic Acids. Res. 15:6625-6641).
  • the antisense nucleic acid molecule can also comprise a 2′-o-methylribonucleotide (Inoue et al. (1987) Nucleic Acids Res. 15:6131-6148) or a chimeric RNA-DNA analogue (Inoue et al. (1987) FEBS Lett. 215:327-330).
  • an antisense nucleic acid of the invention is a ribozyme.
  • Ribozymes are catalytic RNA molecules with ribonuclease activity which are capable of cleaving a single-stranded nucleic acid, such as an mRNA, to which they have a complementary region.
  • ribozymes e.g., hammerhead ribozymes (described in Haselhoff and Gerlach (1988) Nature 334:585-591)) can be used to catalytically cleave MR mRNA transcripts to thereby inhibit translation of MR mRNA.
  • a ribozyme having specificity for an MR-encoding nucleic acid can be designed based upon the nucleotide sequence of an MR DNA disclosed herein (i.e., SEQ ID NO:1 (RXN03181 in Appendix A)).
  • SEQ ID NO:1 RXN03181 in Appendix A
  • a derivative of a Tetrahymena L-19 IVS RNA can be constructed in which the nucleotide sequence of the active site is complementary to the nucleotide sequence to be cleaved in an MR-encoding mRNA. See, e.g., Cech et al. U.S. Pat. No. 4,987,071 and Cech et al. U.S. Pat. No. 5,116,742.
  • MR mRNA can be used to select a catalytic RNA having a specific ribonuclease activity from a pool of RNA molecules. See, e.g., Bartel, D. and Szostak, J. W. (1993) Science 261:1411-1418.
  • MR gene expression can be inhibited by targeting nucleotide sequences complementary to the regulatory region of an MR nucleotide sequence (e.g., an MR promoter and/or enhancers) to form triple helical structures that prevent transcription of an MR gene in target cells.
  • an MR nucleotide sequence e.g., an MR promoter and/or enhancers
  • MR gene expression can be inhibited by targeting nucleotide sequences complementary to the regulatory region of an MR nucleotide sequence (e.g., an MR promoter and/or enhancers) to form triple helical structures that prevent transcription of an MR gene in target cells.
  • nucleotide sequences complementary to the regulatory region of an MR nucleotide sequence e.g., an MR promoter and/or enhancers
  • vectors preferably expression vectors, containing a nucleic acid encoding an MR protein (or a portion thereof).
  • vector refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • plasmid refers to a circular double stranded DNA loop into which additional DNA segments can be ligated.
  • viral vector Another type of vector is a viral vector, wherein additional DNA segments can be ligated into the viral genome.
  • Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors).
  • vectors e.g., non-episomal mammalian vectors
  • Other vectors are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome.
  • certain vectors are capable of directing the expression of genes to which they are operatively linked.
  • Such vectors are referred to herein as “expression vectors”.
  • expression vectors of utility in recombinant DNA techniques are often in the form of plasmids.
  • plasmid and vector can be used interchangeably as the plasmid is the most commonly used form of vector.
  • the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions.
  • the recombinant expression vectors of the invention comprise a nucleic acid of the invention in a form suitable for expression of the nucleic acid in a host cell, which means that the recombinant expression vectors include one or more regulatory sequences, selected on the basis of the host cells to be used for expression, which is operatively linked to the nucleic acid sequence to be expressed.
  • “operably linked” is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner which allows for expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell).
  • regulatory sequence is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel; Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990). Regulatory sequences include those which direct constitutive expression of a nucleotide sequence in many types of host cell and those which direct expression of the nucleotide sequence only in certain host cells.
  • Preferred regulatory sequences are, for example, promoters such as cos-, tac-, trp-, tet-, trp-tet-, lpp-, lac-, lpp-lac-, lacI q -, T7-, T5-, T3-, gal-, trc-, ara-, SP6-, arny, SPO2, ⁇ -P R - or ⁇ P L , which are used preferably in bacteria.
  • promoters such as cos-, tac-, trp-, tet-, trp-tet-, lpp-, lac-, lpp-lac-, lacI q -, T7-, T5-, T3-, gal-, trc-, ara-, SP6-, arny, SPO2, ⁇ -P R - or ⁇ P L , which are used preferably in bacteria.
  • Additional regulatory sequences are, for example, promoters from yeasts and fungi, such as ADC1, MF ⁇ , AC, P-60, CYC1, GAPDH, TEF, rp28, ADH, promoters from plants such as CaMV/35S, SSU, OCS, lib4, usp, STLS1, B33, nos or ubiquitin- or phaseolin-promoters. It is also possible to use artificial promoters. It will be appreciated by one of ordinary skill in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc.
  • the expression vectors of the invention can be introduced into host cells to thereby produce proteins or peptides, including fusion proteins or peptides, encoded by nucleic acids as described herein (e.g., MR proteins, mutant forms of MR proteins, fusion proteins, etc.).
  • the recombinant expression vectors of the invention can be designed for expression of MR proteins in prokaryotic or eukaryotic cells.
  • MR genes can be expressed in bacterial cells such as C. glutamicum , insect cells (using baculovirus expression vectors), yeast and other fungal cells (see Romanos, M. A. et al. (1992) “Foreign gene expression in yeast: a review”, Yeast 8: 423-488; van den Hondel, C. A. M. J. J. et al. (1991) “Heterologous gene expression in filamentous fungi” in: More Gene Manipulations in Fungi, J. W. Bennet & L. L. Lasure, eds., p.
  • telomeres Suitable host cells are discussed further in Goeddel, Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990).
  • the recombinant expression vector can be transcribed and translated in vitro, for example using T7 promoter regulatory sequences and T7 polymerase.
  • Fusion vectors add a number of amino acids to a protein encoded therein, usually to the amino terminus of the recombinant protein.
  • Such fusion vectors typically serve three purposes: 1) to increase expression of recombinant protein; 2) to increase the solubility of the recombinant protein; and 3) to aid in the purification of the recombinant protein by acting as a ligand in affinity purification.
  • a proteolytic cleavage site is introduced at the junction of the fusion moiety and the recombinant protein to enable separation of the recombinant protein from the fusion moiety subsequent to purification of the fusion protein.
  • enzymes, and their cognate recognition sequences include Factor Xa, thrombin and enterokinase.
  • Typical fusion expression vectors include pGEX (Pharmacia Biotech Inc; Smith, D. B. and Johnson, K. S. (1988) Gene 67:31-40), pMAL (New England Biolabs, Beverly, Mass.) and pRIT5 (Pharmacia, Piscataway, N.J.) which fuse glutathione S-transferase (GST), maltose E binding protein, or protein A, respectively, to the target recombinant protein.
  • GST glutathione S-transferase
  • maltose E binding protein or protein A, respectively
  • the coding sequence of the MR protein is cloned into a pGEX expression vector to create a vector encoding a fusion protein comprising, from the N-terminus to the C-terminus, GST-thrombin cleavage site-X protein.
  • the fusion protein can be purified by affinity chromatography using glutathione-agarose resin. Recombinant MR protein unfused to GST can be recovered by cleavage of the fusion protein with thrombin.
  • Suitable inducible non-fusion E. coli expression vectors include pTrc (Amann et al., (1988) Gene 69:301-315) pLG338, pACYC184, pBR322, pUC18, pUC19, pKC30, pRep4, pHS1, pHS2, pPLc236, pMBL24, pLG200, pUR290, pIN-III113-B1, ⁇ gt11, pBdCl, and pET 11d (Studier et al., Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990) 60-89; and Pouwels et al., eds.
  • Target gene expression from the pTrc vector relies on host RNA polymerase transcription from a hybrid trp-lac fusion promoter.
  • Target gene expression from the pET 11d vector relies on transcription from a T7 gn10-lac fusion promoter mediated by a coexpressed viral RNA polymerase (T7 gn1). This viral polymerase is supplied by host strains BL21(DE3) or HMS174(DE3) from a resident ⁇ prophage harboring a T7 gn1 gene under the transcriptional control of the lacUV 5 promoter. For transformation of other varieties of bacteria, appropriate vectors may be selected.
  • the plasmids pIJ101, pIJ364, pIJ702 and pIJ361 are known to be useful in transforming Streptomyces
  • plasmids pUB110, pC194, or pBD214 are suited for transformation of Bacillus species.
  • plasmids of use in the transfer of genetic information into Corynebacterium include pHM1519, pBL1, pSA77, or pAJ667 (Pouwels et al., eds. (1985) Cloning Vectors. Elsevier: New York IBSN 0 444 904018).
  • One strategy to maximize recombinant protein expression is to express the protein in a host bacteria with an impaired capacity to proteolytically cleave the recombinant protein (Gottesman, S., Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990) 119-128).
  • Another strategy is to alter the nucleic acid sequence of the nucleic acid to be inserted into an expression vector so that the individual codons for each amino acid are those preferentially utilized in the bacterium chosen for expression, such as C. glutamicum (Wada et al. (1992) Nucleic Acids Res. 20:2111-2118).
  • Such alteration of nucleic acid sequences of the invention can be carried out by standard DNA synthesis techniques.
  • the MR protein expression vector is a yeast expression vector.
  • yeast expression vectors for expression in yeast S. cerevisiae include pYepSec1 (Baldari, et al., (1987) Embo J. 6:229-234), 2 ⁇ , pAG-1, Yep6, Yep13, pEMBLYe23, pMFa (Kurjan and Herskowitz, (1982) Cell 30:933-943), pJRY88 (Schultz et al., (1987) Gene 54:113-123), and pYES2 (Invitrogen Corporation, San Diego, Calif.).
  • Vectors and methods for the construction of vectors appropriate for use in other fungi, such as the filamentous fungi include those detailed in: van den Hondel, C. A. M. J. J. & Punt, P. J. (1991) “Gene transfer systems and vector development for filamentous fungi, in: Applied Molecular Genetics of Fungi, J. F. Peberdy, et al., eds., p. 1-28, Cambridge University Press: Cambridge, and Pouwels et al., eds. (1985) Cloning Vectors. Elsevier: New York (IBSN 0 444 904018).
  • the MR proteins of the invention can be expressed in insect cells using baculovirus expression vectors.
  • Baculovirus vectors available for expression of proteins in cultured insect cells include the pAc series (Smith et al. (1983) Mol. Cell Biol. 3:2156-2165) and the pVL series (Lucklow and Summers (1989) Virology 170:31-39).
  • the MR proteins of the invention may be expressed in unicellular plant cells (such as algae) or in plant cells from higher plants (e.g., the spermatophytes, such as crop plants).
  • plant expression vectors include those detailed in: Becker, D., Kemper, E., Schell, J. and Masterson, R. (1992) “New plant binary vectors with selectable markers located proximal to the left border”, Plant Mol. Biol. 20: 1195-1197; and Bevan, M. W. (1984) “Binary Agrobacterium vectors for plant transformation”, Nucl. Acid. Res.
  • a nucleic acid of the invention is expressed in mammalian cells using a mammalian expression vector.
  • mammalian expression vectors include pCDM8 (Seed, B. (1987) Nature 329:840) and pMT2PC (Kaufman et al. (1987) EMBO J. 6:187-195).
  • the expression vector's control functions are often provided by viral regulatory elements.
  • commonly used promoters are derived from polyoma, Adenovirus 2, cytomegalovirus and Simian Virus 40.
  • suitable expression systems for both prokaryotic and eukaryotic cells see chapters 16 and 17 of Sambrook, J., Fritsh, E. F., and Maniatis, T. Molecular Cloning: A Laboratory Manual. 2 nd, ed ., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989.
  • the recombinant mammalian expression vector is capable of directing expression of the nucleic acid preferentially in a particular cell type (e.g., tissue-specific regulatory elements are used to express the nucleic acid).
  • tissue-specific regulatory elements are known in the art.
  • suitable tissue-specific promoters include the albumin promoter (liver-specific; Pinkert et al. (1987) Genes Dev. 1:268-277), lymphoid-specific promoters (Calame and Eaton (1988) Adv. Immunol. 43:235-275), in particular promoters of T cell receptors (Winoto and Baltimore (1989) EMBO J.
  • promoters are also encompassed, for example the murine hox promoters (Kessel and Gruss (1990) Science 249:374-379) and the ⁇ -fetoprotein promoter (Campes and Tilghman (1989) Genes Dev. 3:537-546).
  • the invention further provides a recombinant expression vector comprising a DNA molecule of the invention cloned into the expression vector in an antisense orientation. That is, the DNA molecule is operatively linked to a regulatory sequence in a manner which allows for expression (by transcription of the DNA molecule) of an RNA molecule which is antisense to MR mRNA. Regulatory sequences operatively linked to a nucleic acid cloned in the antisense orientation can be chosen which direct the continuous expression of the antisense RNA molecule in a variety of cell types, for instance viral promoters and/or enhancers, or regulatory sequences can be chosen which direct constitutive, tissue specific or cell type specific expression of antisense RNA.
  • the antisense expression vector can be in the form of a recombinant plasmid, phagemid or attenuated virus in which antisense nucleic acids are produced under the control of a high efficiency regulatory region, the activity of which can be determined by the cell type into which the vector is introduced.
  • a high efficiency regulatory region the activity of which can be determined by the cell type into which the vector is introduced.
  • host cell and “recombinant host cell” are used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
  • a host cell can be any prokaryotic or eukaryotic cell.
  • an MR protein can be expressed in bacterial cells such as C. glutamicum , insect cells, yeast or mammalian cells (such as Chinese hamster ovary cells (CHO) or COS cells).
  • suitable host cells are known to one of ordinary skill in the art.
  • Microorganisms related to Corynebacterium glutamicum which may be conveniently used as host cells for the nucleic acid and protein molecules of the invention are set forth in Table 3.
  • Vector DNA can be introduced into prokaryotic or eukaryotic cells via conventional transformation or transfection techniques.
  • transformation and “transfection” are intended to refer to a variety of art-recognized techniques for introducing foreign nucleic acid (e.g., linear DNA or RNA (e.g., a linearized vector or a gene construct alone without a vector) or nucleic acid in the form of a vector (e.g., a plasmid, phage, phasmid, phagemid, transposon or other DNA) into a host cell, including calcium phosphate or calcium chloride co-precipitation, DEAE-dextran-mediated transfection, lipofection, or electroporation.
  • foreign nucleic acid e.g., linear DNA or RNA (e.g., a linearized vector or a gene construct alone without a vector) or nucleic acid in the form of a vector (e.g., a plasmid, phage, phasmid,
  • Suitable methods for transforming or transfecting host cells can be found in Sambrook, et al. ( Molecular Cloning: A Laboratory Manual. 2 nd, ed ., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989), and other laboratory manuals.
  • a gene that encodes a selectable marker (e.g., resistance to antibiotics) is generally introduced into the host cells along with the gene of interest.
  • selectable markers include those which confer resistance to drugs, such as G418, hygromycin and methotrexate.
  • Nucleic acid encoding a selectable marker can be introduced into a host cell on the same vector as that encoding an MR protein or can be introduced on a separate vector. Cells stably transfected with the introduced nucleic acid can be identified by, for example, drug selection (e.g., cells that have incorporated the selectable marker gene will survive, while the other cells die).
  • a vector which contains at least a portion of an MR gene into which a deletion, addition or substitution has been introduced to thereby alter, e.g., functionally disrupt, the MR gene.
  • this MR gene is a Corynebacterium glutamicum MR gene, but it can be a homologue from a related bacterium or even from a mammalian, yeast, or insect source.
  • the vector is designed such that, upon homologous recombination, the endogenous MR gene is functionally disrupted (i.e., no longer encodes a functional protein; also referred to as a “knock out” vector).
  • the vector can be designed such that, upon homologous recombination, the endogenous MR gene is mutated or otherwise altered but still encodes functional protein (e.g., the upstream regulatory region can be altered to thereby alter the expression of the endogenous MR protein).
  • the altered portion of the MR gene is flanked at its 5′ and 3′ ends by additional nucleic acid of the MR gene to allow for homologous recombination to occur between the exogenous MR gene carried by the vector and an endogenous MR gene in a microorganism.
  • the additional flanking MR nucleic acid is of sufficient length for successful homologous recombination with the endogenous gene.
  • flanking DNA both at the 5′ and 3′ ends
  • flanking DNA both at the 5′ and 3′ ends
  • the vector is introduced into a microorganism (e.g., by electroporation) and cells in which the introduced MR gene has homologously recombined with the endogenous MR gene are selected, using art-known techniques.
  • recombinant microorganisms can be produced which contain selected systems which allow for regulated expression of the introduced gene.
  • inclusion of an MR gene on a vector placing it under control of the lac operon permits expression of the MR gene only in the presence of IPTG.
  • Such regulatory systems are well known in the art.
  • an endogenous MR gene in a host cell is disrupted (e.g., by homologous recombination or other genetic means known in the art) such that expression of its protein product does not occur.
  • an endogenous or introduced MR gene in a host cell has been altered by one or more point mutations, deletions, or inversions, but still encodes a functional MR protein.
  • one or more of the regulatory regions (e.g., a promoter, repressor, or inducer) of an MR gene in a microorganism has been altered (e.g., by deletion, truncation, inversion, or point mutation) such that the expression of the MR gene is modulated.
  • a host cell of the invention such as a prokaryotic or eukaryotic host cell in culture, can be used to produce (i.e., express) an MR protein.
  • the invention further provides methods for producing MR proteins using the host cells of the invention.
  • the method comprises culturing the host cell of invention (into which a recombinant expression vector encoding an MR protein has been introduced, or into which genome has been introduced a gene encoding a wild-type or altered MR protein) in a suitable medium until MR protein is produced.
  • the method further comprises isolating MR proteins from the medium or the host cell.
  • Another aspect of the invention pertains to isolated MR proteins, and biologically active portions thereof.
  • An “isolated” or “purified” protein or biologically active portion thereof is substantially free of cellular material when produced by recombinant DNA techniques, or chemical precursors or other chemicals when chemically synthesized.
  • the language “substantially free of cellular material” includes preparations of MR protein in which the protein is separated from cellular components of the cells in which it is naturally or recombinantly produced.
  • the language “substantially free of cellular material” includes preparations of MR protein having less than about 30% (by dry weight) of non-MR protein (also referred to herein as a “contaminating protein”), more preferably less than about 20% of non-MR protein, still more preferably less than about 10% of non-MR protein, and most preferably less than about 5% non-MR protein.
  • non-MR protein also referred to herein as a “contaminating protein”
  • contaminating protein also preferably substantially free of culture medium, i.e., culture medium represents less than about 20%, more preferably less than about 10%, and most preferably less than about 5% of the volume of the protein preparation.
  • the language “substantially free of chemical precursors or other chemicals” includes preparations of MR protein in which the protein is separated from chemical precursors or other chemicals which are involved in the synthesis of the protein.
  • the language “substantially free of chemical precursors or other chemicals” includes preparations of MR protein having less than about 30% (by dry weight) of chemical precursors or non-MR chemicals, more preferably less than about 20% chemical precursors or non-MR chemicals, still more preferably less than about 10% chemical precursors or non-MR chemicals, and most preferably less than about 5% chemical precursors or non-MR chemicals.
  • isolated proteins or biologically active portions thereof lack contaminating proteins from the same organism from which the MR protein is derived. Typically, such proteins are produced by recombinant expression of, for example, a C. glutamicum MR protein in a microorganism such as C. glutamicum.
  • an isolated MR protein or a portion thereof of the invention can transcriptionally, translationally, or posttranslationally regulate a metabolic pathway in C. glutamicum , or has one or more of the activities set forth in Table 1.
  • the protein or portion thereof comprises an amino acid sequence which is sufficiently homologous to an amino acid sequence of Appendix B such that the protein or portion thereof maintains the ability to transcriptionally, translationally, or posttranslationally regulate a metabolic pathway in C. glutamicum .
  • the portion of the protein is preferably a biologically active portion as described herein.
  • an MR protein of the invention has an amino acid sequence shown in Appendix B.
  • the MR protein has an amino acid sequence which is encoded by a nucleotide sequence which hybridizes, e.g., hybridizes under stringent conditions, to a nucleotide sequence of Appendix A.
  • the MR protein has an amino acid sequence which is encoded by a nucleotide sequence that is at least about 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, or 60%, preferably at least about 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, or 70%, more preferably at least about 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, or 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, or 90%, or 91%, 92%, 93%, 94%, and even more preferably at least about 95%, 96%
  • Ranges and identity values intermediate to the above-recited values, are also intended to be encompassed by the present invention.
  • ranges of identity values using a combination of any of the above values recited as upper and/or lower limits are intended to be included.
  • the preferred MR proteins of the present invention also preferably possess at least one of the MR activities described herein.
  • a preferred MR protein of the present invention includes an amino acid sequence encoded by a nucleotide sequence which hybridizes, e.g., hybridizes under stringent conditions, to a nucleotide sequence of Appendix A, and which can transcriptionally, translationally, or posttranslationally regulate a metabolic pathway in C. glutamicum , or which has one or more of the activities set forth in Table 1.
  • the MR protein is substantially homologous to an amino acid sequence of Appendix B and retains the functional activity of the protein of one of the sequences of Appendix B yet differs in amino acid sequence due to natural variation or mutagenesis, as described in detail in subsection I above.
  • the MR protein is a protein which comprises an amino acid sequence which is at least about 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, or 60%, preferably at least about 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, or 70%, more preferably at least about 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, or 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, or 90%, or 91%, 92%, 93%, 94%, and even more preferably at least about 95%, 96%, 97%, 98%, 99% or more homologous to an entire amino acid sequence of Appendix B and which has at least one of the MR activities described herein.
  • Ranges and identity values intermediate to the above-recited values are also intended to be encompassed by the present invention.
  • ranges of identity values using a combination of any of the above values recited as upper and/or lower limits are intended to be included.
  • the invention pertains to a full length C. glutamicum protein which is substantially homologous to an entire amino acid sequence of Appendix B.
  • Biologically active portions of an MR protein include peptides comprising amino acid sequences derived from the amino acid sequence of an MR protein, e.g., the an amino acid sequence shown in Appendix B or the amino acid sequence of a protein homologous to an MR protein, which include fewer amino acids than a full length MR protein or the full length protein which is homologous to an MR protein, and exhibit at least one activity of an MR protein.
  • biologically active portions peptides, e.g., peptides which are, for example, 5, 10, 15, 20, 30, 35, 36, 37, 38, 39, 40, 50, 100 or more amino acids in length
  • biologically active portions in which other regions of the protein are deleted, can be prepared by recombinant techniques and evaluated for one or more of the activities described herein.
  • the biologically active portions of an MR protein include one or more selected domains/motifs or portions thereof having biological activity.
  • MR proteins are preferably produced by recombinant DNA techniques.
  • a nucleic acid molecule encoding the protein is cloned into an expression vector (as described above), the expression vector is introduced into a host cell (as described above) and the MR protein is expressed in the host cell.
  • the MR protein can then be isolated from the cells by an appropriate purification scheme using standard protein purification techniques.
  • an MR protein, polypeptide, or peptide can be synthesized chemically using standard peptide synthesis techniques.
  • native MR protein can be isolated from cells (e.g., endothelial cells), for example using an anti-MR antibody, which can be produced by standard techniques utilizing an MR protein or fragment thereof of this invention.
  • an MR “chimeric protein” or “fusion protein” comprises an MR polypeptide operatively linked to a non-MR polypeptide.
  • An “MR polypeptide” refers to a polypeptide having an amino acid sequence corresponding to an MR protein
  • a “non-MR polypeptide” refers to a polypeptide having an amino acid sequence corresponding to a protein which is not substantially homologous to the MR protein, e.g., a protein which is different from the MR protein and which is derived from the same or a different organism.
  • the term “operatively linked” is intended to indicate that the MR polypeptide and the non-MR polypeptide are fused in-frame to each other.
  • the non-MR polypeptide can be fused to the N-terminus or C-terminus of the MR polypeptide.
  • the fusion protein is a GST-MR fusion protein in which the MR sequences are fused to the C-terminus of the GST sequences.
  • Such fusion proteins can facilitate the purification of recombinant MR proteins.
  • the fusion protein is an MR protein containing a heterologous signal sequence at its N-terminus. In certain host cells (e.g., mammalian host cells), expression and/or secretion of an MR protein can be increased through use of a heterologous signal sequence.
  • an MR chimeric or fusion protein of the invention is produced by standard recombinant DNA techniques.
  • DNA fragments coding for the different polypeptide sequences are ligated together in-frame in accordance with conventional techniques, for example by employing blunt-ended or stagger-ended termini for ligation, restriction enzyme digestion to provide for appropriate termini, filling-in of cohesive ends as appropriate, alkaline phosphatase treatment to avoid undesirable joining, and enzymatic ligation.
  • the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers.
  • PCR amplification of gene fragments can be carried out using anchor primers which give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed and reamplified to generate a chimeric gene sequence (see, for example, Current Protocols in Molecular Biology , eds. Ausubel et al. John Wiley & Sons: 1992).
  • anchor primers which give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed and reamplified to generate a chimeric gene sequence
  • many expression vectors are commercially available that already encode a fusion moiety (e.g., a GST polypeptide).
  • An MR-encoding nucleic acid can be cloned into such an expression vector such that the fusion moiety is linked in-frame to the MR protein.
  • Homologues of the MR protein can be generated by mutagenesis, e.g., discrete point mutation or truncation of the MR protein.
  • the term “homologue” refers to a variant form of the MR protein which acts as an agonist or antagonist of the activity of the MR protein.
  • An agonist of the MR protein can retain substantially the same, or a subset, of the biological activities of the MR protein.
  • An antagonist of the MR protein can inhibit one or more of the activities of the naturally occurring form of the MR protein, by, for example, competitively binding to a downstream or upstream member of the MR regulatory cascade which includes the MR protein.
  • the C. glutamicum MR protein and homologues thereof of the present invention may modulate the activity of one or more metabolic pathways which MR proteins regulate in this microorganism.
  • homologues of the MR protein can be identified by screening combinatorial libraries of mutants, e.g., truncation mutants, of the MR protein for MR protein agonist or antagonist activity.
  • a variegated library of MR variants is generated by combinatorial mutagenesis at the nucleic acid level and is encoded by a variegated gene library.
  • a variegated library of MR variants can be produced by, for example, enzymatically ligating a mixture of synthetic oligonucleotides into gene sequences such that a degenerate set of potential MR sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g., for phage display) containing the set of MR sequences therein.
  • a degenerate set of potential MR sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g., for phage display) containing the set of MR sequences therein.
  • fusion proteins e.g., for phage display
  • degenerate set of genes allows for the provision, in one mixture, of all of the sequences encoding the desired set of potential MR sequences.
  • Methods for synthesizing degenerate oligonucleotides are known in the art (see, e.g., Narang, S. A. (1983) Tetrahedron 39:3; Itakura et al. (1984) Annu. Rev. Biochem. 53:323; Itakura et al. (1984) Science 198:1056; Ike et al. (1983) Nucleic Acid Res. 11:477.
  • libraries of fragments of the MR protein coding can be used to generate a variegated population of MR fragments for screening and subsequent selection of homologues of an MR protein.
  • a library of coding sequence fragments can be generated by treating a double stranded PCR fragment of an MR coding sequence with a nuclease under conditions wherein nicking occurs only about once per molecule, denaturing the double stranded DNA, renaturing the DNA to form double stranded DNA which can include sense/antisense pairs from different nicked products, removing single stranded portions from reformed duplexes by treatment with S1 nuclease, and ligating the resulting fragment library into an expression vector.
  • an expression library can be derived which encodes N-terminal, C-terminal and internal fragments of various sizes of the MR protein.
  • REM Recursive ensemble mutagenesis
  • cell based assays can be exploited to analyze a variegated MR library, using methods well known in the art.
  • nucleic acid molecules, proteins, protein homologues, fusion proteins, primers, vectors, and host cells described herein can be used in one or more of the following methods: identification of C. glutamicum and related organisms; mapping of genomes of organisms related to C. glutamicum ; identification and localization of C. glutamicum sequences of interest; evolutionary studies; determination of MR protein regions required for function; modulation of an MR protein activity; modulation of the activity of one or more metabolic pathways; and modulation of cellular production of a desired compound, such as a fine chemical.
  • the MR nucleic acid molecules of the invention have a variety of uses. First, they may be used to identify an organism as being Corynebacterium glutamicum or a close relative thereof. Also, they may be used to identify the presence of C. glutamicum or a relative thereof in a mixed population of microorganisms.
  • the invention provides the nucleic acid sequences of a number of C. glutamicum genes; by probing the extracted genomic DNA of a culture of a unique or mixed population of microorganisms under stringent conditions with a probe spanning a region of a C. glutamicum gene which is unique to this organism, one can ascertain whether this organism is present.
  • Corynebacterium glutamicum itself is nonpathogenic, it is related to pathogenic species, such as Corynebacterium diphtheriae.
  • Corynebacterium diphtheriae is the causative agent of diphtheria, a rapidly developing, acute, febrile infection which involves both local and systemic pathology.
  • a local lesion develops in the upper respiratory tract and involves necrotic injury to epithelial cells; the bacilli secrete toxin which is disseminated through this lesion to distal susceptible tissues of the body.
  • Degenerative changes brought about by the inhibition of protein synthesis in these tissues which include heart, muscle, peripheral nerves, adrenals, kidneys, liver and spleen, result in the systemic pathology of the disease.
  • Diphtheria continues to have high incidence in many parts of the world, including Africa, Asia, Eastern Europe and the independent states of the former Soviet Union. An ongoing epidemic of diphtheria in the latter two regions has resulted in at least 5,000 deaths since 1990.
  • the invention provides a method of identifying the presence or activity of Cornyebacterium diphtheriae in a subject.
  • This method includes detection of one or more of the nucleic acid or amino acid sequences of the invention (e.g., the sequences set forth in Appendix A or Appendix B) in a subject, thereby detecting the presence or activity of Corynebacterium diphtheriae in the subject.
  • C. glutamicum and C. diphtheriae are related bacteria, and many of the nucleic acid and protein molecules in C. glutamicum are homologous to C. diphtheriae nucleic acid and protein molecules, and can therefore be used to detect C. diphtheriae in a subject.
  • the nucleic acid and protein molecules of the invention may also serve as markers for specific regions of the genome. This has utility not only in the mapping of the genome, but also for functional studies of C. glutamicum proteins. For example, to identify the region of the genome to which a particular C. glutamicum DNA-binding protein binds, the C. glutamicum genome could be digested, and the fragments incubated with the DNA-binding protein. Those which bind the protein may be additionally probed with the nucleic acid molecules of the invention, preferably with readily detectable labels; binding of such a nucleic acid molecule to the genome fragment enables the localization of the fragment to the genome map of C.
  • nucleic acid molecules of the invention may be sufficiently homologous to the sequences of related species such that these nucleic acid molecules may serve as markers for the construction of a genomic map in related bacteria, such as Brevibacterium lactofermentum.
  • the MR nucleic acid molecules of the invention are also useful for evolutionary and protein structural studies.
  • the metabolic processes in which the molecules of the invention participate are utilized by a wide variety of prokaryotic and eukaryotic cells; by comparing the sequences of the nucleic acid molecules of the present invention to those encoding similar enzymes from other organisms, the evolutionary relatedness of the organisms can be assessed. Similarly, such a comparison permits an assessment of which regions of the sequence are conserved and which are not, which may aid in determining those regions of the protein which are essential for the functioning of the enzyme. This type of determination is of value for protein engineering studies and may give an indication of what the protein can tolerate in terms of mutagenesis without losing function.
  • Manipulation of the MR nucleic acid molecules of the invention may result in the production of MR proteins having functional differences from the wild-type MR proteins. These proteins may be improved in efficiency or activity, may be present in greater numbers in the cell than is usual, or may be decreased in efficiency or activity.
  • the invention provides methods for screening molecules which modulate the activity of an MR protein, either by interacting with the protein itself or a substrate or binding partner of the MR protein, or by modulating the transcription or translation of an MR nucleic acid molecule of the invention.
  • a microorganism expressing one or more MR proteins of the invention is contacted with one or more test compounds, and the effect of each test compound on the activity or level of expression of the MR protein is assessed.
  • Such changes in activity may directly modulate the yield, production, and/or efficiency of production of one or more fine chemicals from C. glutamicum .
  • MR protein which activates the transcription or translation of a gene encoding a biosynthetic protein for a desired fine chemical
  • impairing or abrogating the activity of an MR protein which represses the transcription or translation of such a gene one may also increase the activity or rate of activity of that biosynthetic pathway due to the presence of increased levels of what may have been a limiting enzyme.
  • an MR protein by altering the activity of an MR protein such that it constitutively posttranslationally inactivates a protein involved in a degradation pathway for a desired fine chemical, or by altering the activity of an MR protein such that it constitutively represses the transcription or translation of such a gene, one may increase the yield and/or rate of production of the fine chemical from the cell, due to decreased degradation of the compound.
  • an MR protein which represses the synthesis of molecules necessary for cell membrane production in response to high levels of waste products in the extracellular medium (in order to block cell growth and division in suboptimal growth conditions) such that it no longer is able to repress such synthesis, one may increase the growth and multiplication of the cell in cultures even when the growth conditions are suboptimal.
  • Such enhanced growth or viability should also increase the yields and/or rate of production of a desired fine chemical from fermentative culture, due to the relatively greater number of cells producing this compound in the culture.
  • the nucleic acid and protein molecules of the invention may be utilized to generate C. glutamicum or related strains of bacteria expressing mutated MR nucleic acid and protein molecules such that the yield and/or efficiency of production of a desired compound is improved.
  • This desired compound may be any natural product of C. glutamicum , which includes the final products of biosynthesis pathways and intermediates of naturally-occurring metabolic pathways, as well as molecules which do not naturally occur in the metabolism of C. glutamicum , but which are produced by a C. glutamicum strain of the invention.
  • a culture of Corynebacterium glutamicum (ATCC 13032) was grown overnight at 30° C. with vigorous shaking in BHI medium (Difco). The cells were harvested by centrifugation, the supernatant was discarded and the cells were resuspended in 5 ml buffer-I (5% of the original volume of the culture—all indicated volumes have been calculated for 100 ml of culture volume).
  • composition of buffer-I 140.34 g/l sucrose, 2.46 g/l MgSO 4 ⁇ 7H 2 O, 10 ml/l KH 2 PO 4 solution (100 g/l, adjusted to pH 6.7 with KOH), 50 ml/l M12 concentrate (10 g/l (NH 4 ) 2 SO 4 , 1 g/l NaCl, 2 ⁇ l MgSO 4 ⁇ 7H 2 O, 0.2 g/l CaCl 2 , 0.5 g/l yeast extract (Difco), 10 ml/l trace-elements-mix (200 mg/l FeSO 4 ⁇ H 2 O, 10 mg/l ZnSO 4 ⁇ 7H 2 O, 3 mg/l MnCl 2 ⁇ 4H 2 O, 30 mg/l H 3 BO 3 20 mg/l CoCl 2 ⁇ 6H 2 O, 1 mg/l NiCl 2 ⁇ 6H 2 O, 3 mg/l Na 2 MoO 4 ⁇ 2H 2 O, 500 mg/l complexing agent (
  • Lysozyme was added to the suspension to a final concentration of 2.5 mg/ml. After an approximately 4 h incubation at 37° C., the cell wall was degraded and the resulting protoplasts are harvested by centrifugation. The pellet was washed once with 5 ml buffer-I and once with 5 ml TE-buffer (10 mM Tris-HCl, 1 mM EDTA, pH 8). The pellet was resuspended in 4 ml TE-buffer and 0.5 ml SDS solution (10%) and 0.5 ml NaCl solution (5 M) are added. After adding of proteinase K to a final concentration of 200 ⁇ g/ml, the suspension is incubated for ca. 18 h at 37° C.
  • the DNA was purified by extraction with phenol, phenol-chloroform-isoamylalcohol and chloroform-isoamylalcohol using standard procedures. Then, the DNA was precipitated by adding 1/50 volume of 3 M sodium acetate and 2 volumes of ethanol, followed by a 30 min incubation at ⁇ 20° C. and a 30 min centrifugation at 12,000 rpm in a high speed centrifuge using a SS34 rotor (Sorvall). The DNA was dissolved in 1 ml TE-buffer containing 20 ⁇ g/ml RNaseA and dialysed at 4° C. against 1000 ml TE-buffer for at least 3 hours. During this time, the buffer was exchanged 3 times.
  • plasmids pBR322 Suddens, J. G. (1979) Proc. Natl. Acad. Sci. USA, 75:3737-3741); pACYC177 (Change & Cohen (1978) J. Bacteriol 134:1141-1156), plasmids of the pBS series (pBSSK+, pBSSK ⁇ and others; Stratagene, LaJolla, USA), or cosmids as SuperCos1 (Stratagene, LaJolla, USA) or Lorist6 (Gibson, T. J., Rosenthal A. and Waterson, R. H. (1987) Gene 53:283-286.
  • Gene libraries specifically for use in C. glutamicum may be constructed using plasmid pSL109 (Lee, H.-S. and A. J. Sinskey (1994) J. Microbiol. Biotechnol. 4: 256-263).
  • Genomic libraries as described in Example 2 were used for DNA sequencing according to standard methods, in particular by the chain termination method using ABI377 sequencing machines (see e.g., Fleischman, R. D. et al. (1995) “Whole-genome Random Sequencing and Assembly of Haemophilus Influenzae Rd., Science, 269:496-512). Sequencing primers with the following nucleotide sequences were used: 5′-GGAAACAGTATGACCATG-3′ or 5′-GTAAAACGACGGCCAGT-3′.
  • In vivo mutagenesis of Corynebacterium glutamicum can be performed by passage of plasmid (or other vector) DNA through E. coli or other microorganisms (e.g. Bacillus spp. or yeasts such as Saccharomyces cerevisiae ) which are impaired in their capabilities to maintain the integrity of their genetic information.
  • E. coli or other microorganisms e.g. Bacillus spp. or yeasts such as Saccharomyces cerevisiae
  • Typical mutator strains have mutations in the genes for the DNA repair system (e.g., mutHLS, mutD, mutT, etc.; for reference, see Rupp, W. D. (1996) DNA repair mechanisms, in: Escherichia coli and Salmonella , p. 2277-2294, ASM: Washington.)
  • Such strains are well known to one of ordinary skill in the art. The use of such strains is illustrated, for example, in Greener, A. and Call
  • Corynebacterium and Brevibacterium species contain endogenous plasmids (as e.g., pHM1519 or pBL1) which replicate autonomously (for review see, e.g., Martin, J. F. et al. (1987) Biotechnology, 5:137-146).
  • Shuttle vectors for Escherichia coli and Corynebacterium glutamicum can be readily constructed by using standard vectors for E. coli (Sambrook, J. et al. (1989), “Molecular Cloning: A Laboratory Manual”, Cold Spring Harbor Laboratory Press or Ausubel, F. M. et al.
  • origins of replication are preferably taken from endogenous plasmids isolated from Corynebacterium and Brevibacterium species.
  • transformation markers are genes for kanamycin resistance (such as those derived from the Tn5 or Tn903 transposons) or chloramphenicol (Winnacker, E. L. (1987) “From Genes to Clones—Introduction to Gene Technology, VCH, Weinheim).
  • kanamycin resistance such as those derived from the Tn5 or Tn903 transposons
  • chloramphenicol Winnacker, E. L. (1987) “From Genes to Clones—Introduction to Gene Technology, VCH, Weinheim.
  • glutamicum which can be used for several purposes, including gene over-expression (for reference, see e.g., Yoshihama, M. et al. (1985) J. Bacteriol. 162:591-597, Martin J. F. et al. (1987) Biotechnology, 5:137-146 and Eikmanns, B. J. et al. (1991) Gene, 102:93-98).
  • Genes may be overexpressed in C. glutamicum strains using plasmids which comprise pCG1 (U.S. Pat. No. 4,617,267) or fragments thereof, and optionally the gene for kanamycin resistance from TN903 (Grindley, N. D. and Joyce, C. M. (1980) Proc. Natl. Acad. Sci. USA 77(12): 7176-7180).
  • genes may be overexpressed in C. glutamicum strains using plasmid pSL109 (Lee, H.-S. and A. J. Sinskey (1994) J. Microbiol. Biotechnol. 4: 256-263).
  • Genomic integration in C. glutamicum or other Corynebacterium or Brevibacterium species may be accomplished by well-known methods, such as homologous recombination with genomic region(s), restriction endonuclease mediated integration (REMI) (see, e.g., DE Patent 19823834), or through the use of transposons.
  • REMI restriction endonuclease mediated integration
  • a gene of interest by modifying the regulatory regions (e.g., a promoter, a repressor, and/or an enhancer) by sequence modification, insertion, or deletion using site-directed methods (such as homologous recombination) or methods based on random events (such as transposon mutagenesis or REMI).
  • Site-directed methods such as homologous recombination
  • random events such as transposon mutagenesis or REMI.
  • Nucleic acid sequences which function as transcriptional terminators may also be inserted 3′ to the coding region of one or more genes of the invention; such terminators are well-known in the art and are described, for example, in Winnacker, E. L. (1987) From Genes to Clones—Introduction to Gene Technology. VCH: Weinheim.
  • Observations of the activity of a mutated protein in a transformed host cell rely on the fact that the mutant protein is expressed in a similar fashion and in a similar quantity to that of the wild-type protein.
  • a useful method to ascertain the level of transcription of the mutant gene is to perform a Northern blot (for reference see, for example, Ausubel et al.
  • RNA of a culture of the organism is extracted, run on gel, transferred to a stable matrix and incubated with this probe, the binding and quantity of binding of the probe indicates the presence and also the quantity of mRNA for this gene. This information is evidence of the degree of transcription of the mutant gene.
  • a detectable tag usually radioactive or chemiluminescent
  • Corynebacteria are cultured in synthetic or natural growth media.
  • a number of different growth media for Corynebacteria are both well-known and readily available (Lieb et al. (1989) Appl. Microbiol. Biotechnol., 32:205-210; von der Osten et al. (1998) Biotechnology Letters, 11:11-16; Patent DE 4,120,867; Liebl (1992) “The Genus Corynebacterium , in: The Procaryotes, Volume II, Balows, A. et al., eds. Springer-Verlag).
  • These media consist of one or more carbon sources, nitrogen sources, inorganic salts, vitamins and trace elements.
  • Preferred carbon sources are sugars, such as mono-, di-, or polysaccharides.
  • sugars such as mono-, di-, or polysaccharides.
  • glucose, fructose, mannose, galactose, ribose, sorbose, ribulose, lactose, maltose, sucrose, raffinose, starch or cellulose serve as very good carbon sources.
  • sugar to the media via complex compounds such as molasses or other by-products from sugar refinement.
  • Other possible carbon sources are alcohols and organic acids, such as methanol, ethanol, acetic acid or lactic acid.
  • Nitrogen sources are usually organic or inorganic nitrogen compounds, or materials which contain these compounds.
  • Exemplary nitrogen sources include ammonia gas or ammonia salts, such as NH 4 Cl or (NH 4 ) 2 SO 4 , NH 4 OH, nitrates, urea, amino acids or complex nitrogen sources like corn steep liquor, soy bean flour, soy bean protein, yeast extract, meat extract and others.
  • Inorganic salt compounds which may be included in the media include the chloride-, phosphorous- or sulfate-salts of calcium, magnesium, sodium, cobalt, molybdenum, potassium, manganese, zinc, copper and iron.
  • Chelating compounds can be added to the medium to keep the metal ions in solution.
  • Particularly useful chelating compounds include dihydroxyphenols, like catechol or protocatechuate, or organic acids, such as citric acid. It is typical for the media to also contain other growth factors, such as vitamins or growth promoters, examples of which include biotin, riboflavin, thiamin, folic acid, nicotinic acid, pantothenate and pyridoxin.
  • the exact composition of the media compounds depends strongly on the immediate experiment and is individually decided for each specific case. Information about media optimization is available in the textbook “Applied Microbiol. Physiology, A Practical Approach (eds. P. M. Rhodes, P. F. Stanbury, IRL Press (1997) pp. 53-73, ISBN 0 19 963577 3). It is also possible to select growth media from commercial suppliers, like standard 1 (Merck) or BHI (grain heart infusion, DIFCO) or others.
  • All medium components are sterilized, either by heat (20 minutes at 1.5 bar and 121° C.) or by sterile filtration.
  • the components can either be sterilized together or, if necessary, separately. All media components can be present at the beginning of growth, or they can optionally be added continuously or batchwise.
  • the temperature should be in a range between 15° C. and 45° C.
  • the temperature can be kept constant or can be altered during the experiment.
  • the pH of the medium should be in the range of 5 to 8.5, preferably around 7.0, and can be maintained by the addition of buffers to the media.
  • An exemplary buffer for this purpose is a potassium phosphate buffer.
  • Synthetic buffers such as MOPS, HEPES, ACES and others can alternatively or simultaneously be used. It is also possible to maintain a constant culture pH through the addition of NaOH or NH 4 OH during growth. If complex medium components such as yeast extract are utilized, the necessity for additional buffers may be reduced, due to the fact that many complex compounds have high buffer capacities. If a fermentor is utilized for culturing the micro-organisms, the pH can also be controlled using gaseous ammonia.
  • the incubation time is usually in a range from several hours to several days. This time is selected in order to permit the maximal amount of product to accumulate in the broth.
  • the disclosed growth experiments can be carried out in a variety of vessels, such as microtiter plates, glass tubes, glass flasks or glass or metal fermentors of different sizes.
  • the microorganisms should be cultured in microtiter plates, glass tubes or shake flasks, either with or without baffles.
  • 100 ml shake flasks are used, filled with 10% (by volume) of the required growth medium.
  • the flasks should be shaken on a rotary shaker (amplitude 25 mm) using a speed-range of 100-300 rpm. Evaporation losses can be diminished by the maintenance of a humid atmosphere; alternatively, a mathematical correction for evaporation losses should be performed.
  • the medium is inoculated to an OD 600 of O.5-1.5 using cells grown on agar plates, such as CM plates (10 g/l glucose, 2.5 g/l NaCl, 2 g/l urea, 10 g/l polypeptone, 5 g/l yeast extract, 5 g/l meat extract, 22 g/l NaCl, 2 g/l urea, 10 g/l polypeptone, 5 g/l yeast extract, 5 g/l meat extract, 22 g/l agar, pH 6.8 with 2M NaOH) that had been incubated at 30° C. Inoculation of the media is accomplished by either introduction of a saline suspension of C. glutamicum cells from CM plates or addition of a liquid preculture of this bacterium.
  • CM plates 10 g/l glucose, 2.5 g/l NaCl, 2 g/l urea, 10 g/l polypeptone, 5 g/l yeast extract, 5 g/l meat
  • DNA band-shift assays also called gel retardation assays
  • reporter gene assays such as that described in Kolmar, H. et al. (1995) EMBO J. 14: 3895-3904 and references cited therein. Reporter gene test systems are well known and established for applications in both pro- and eukaryotic cells, using enzymes such as beta-galactosidase, green fluorescent protein, and several others.
  • membrane-transport proteins The determination of activity of membrane-transport proteins can be performed according to techniques such as those described in Gennis, R. B. (1989) “Pores, Channels and Transporters”, in Biomembranes, Molecular Structure and Function, Springer: Heidelberg, p. 85-137; 199-234; and 270-322.
  • the effect of the genetic modification in C. glutamicum on production of a desired compound can be assessed by growing the modified microorganism under suitable conditions (such as those described above) and analyzing the medium and/or the cellular component for increased production of the desired product (i.e., an amino acid).
  • suitable conditions such as those described above
  • Such analysis techniques are well known to one of ordinary skill in the art, and include spectroscopy, thin layer chromatography, staining methods of various kinds, enzymatic and microbiological methods, and analytical chromatography such as high performance liquid chromatography (see, for example, Ullman, Encyclopedia of Industrial Chemistry, vol. A2, p. 89-90 and p. 443-613, VCH: Weinheim (1985); Fallon, A.
  • Recovery of the desired product from the C. glutamicum cells or supernatant of the above-described culture can be performed by various methods well known in the art. If the desired product is not secreted from the cells, the cells can be harvested from the culture by low-speed centrifugation, the cells can be lysed by standard techniques, such as mechanical force or sonication. The cellular debris is removed by centrifugation, and the supernatant fraction containing the soluble proteins is retained for further purification of the desired compound. If the product is secreted from the C. glutamicum cells, then the cells are removed from the culture by low-speed centrifugation, and the supernate fraction is retained for further purification.
  • the supernatant fraction from either purification method is subjected to chromatography with a suitable resin, in which the desired molecule is either retained on a chromatography resin while many of the impurities in the sample are not, or where the impurities are retained by the resin while the sample is not.
  • chromatography steps may be repeated as necessary, using the same or different chromatography resins.
  • One of ordinary skill in the art would be well-versed in the selection of appropriate chromatography resins and in their most efficacious application for a particular molecule to be purified.
  • the purified product may be concentrated by filtration or ultrafiltration, and stored at a temperature at which the stability of the product is maximized.
  • the identity and purity of the isolated compounds may be assessed by techniques standard in the art. These include high-performance liquid chromatography (HPLC), spectroscopic methods, staining methods, thin layer chromatography, NIRS, enzymatic assay, or microbiologically. Such analysis methods are reviewed in: Patek et al. (1994) Appl. Environ. Microbiol. 60: 133-140; Malakhova et al. (1996) Biotekhnologiya 11: 27-32; and Schmidt et al. (1998) Bioprocess Engineer. 19: 67-70. Ulmann's Encyclopedia of Industrial Chemistry, (1996) vol. A27, VCH: Weinheim, p. 89-90, p. 521-540, p. 540-547, p.
  • Gapped BLAST can be utilized as described in Altschul et al., (1997) Nucleic Acids Res. 25(17):3389-3402.
  • Another example of a mathematical algorithm utilized for the comparison of sequences is the algorithm of Meyers and Miller ((1988) Comput. Appl. Biosci. 4: 11-17). Such an algorithm is incorporated into the ALIGN program (version 2.0) which is part of the GCG sequence alignment software package. When utilizing the ALIGN program for comparing amino acid sequences, a PAM120 weight residue table, a gap length penalty of 12, and a gap penalty of 4 can be used. Additional algorithms for sequence analysis are known in the art, and include ADVANCE and ADAM. described in Torelli and Robotti (1994) Comput. Appl. Biosci. 10:3-5; and FASTA, described in Pearson and Lipman (1988) P.N.A.S. 85:2444-8.
  • the percent homology between two amino acid sequences can also be accomplished using the GAP program in the GCG software package (available at http://www.gcg.com), using either a Blosum 62 matrix or a PAM250 matrix, and a gap weight of 12, 10, 8, 6, or 4 and a length weight of 2, 3, or 4.
  • the percent homology between two nucleic acid sequences can be accomplished using the GAP program in the GCG software package, using standard parameters, such as a gap weight of 50 and a length weight of 3.
  • a comparative analysis of the gene sequences of the invention with those present in Genbank has been performed using techniques known in the art (see, e.g., Bexevanis and Ouellette, eds. (1998) Bioinformatics: A Practical Guide to the Analysis of Genes and Proteins. John Wiley and Sons: New York).
  • the gene sequences of the invention were compared to genes present in Genbank in a three-step process.
  • a BLASTN analysis e.g., a local alignment analysis
  • a subsequent FASTA search (e.g., a combined local and global alignment analysis, in which limited regions of the sequences are aligned) was performed on these 500 hits.
  • Each gene sequence of the invention was subsequently globally aligned to each of the top three FASTA hits, using the GAP program in the GCG software package (using standard parameters).
  • the length of the sequences extracted from Genbank were adjusted to the length of the query sequences by methods well-known in the art. The results of this analysis are set forth in Table 4.
  • sequences of the invention may additionally be used in the construction and application of DNA microarrays (the design, methodology, and uses of DNA arrays are well known in the art, and are described, for example, in Schena, M. et al. (1995) Science 270: 467-470; Wodicka, L. et al. (1997) Nature Biotechnology 15: 1359-1367; DeSaizieu, A. et al. (1998) Nature Biotechnology 16: 45-48; and DeRisi, J. L. et al. (1997) Science 278: 680-686).
  • DNA microarrays are solid or flexible supports consisting of nitrocellulose, nylon, glass, silicone, or other materials. Nucleic acid molecules may be attached to the surface in an ordered manner. After appropriate labeling, other nucleic acids or nucleic acid mixtures can be hybridized to the immobilized nucleic acid molecules, and the label may be used to monitor and measure the individual signal intensities of the hybridized molecules at defined regions. This methodology allows the simultaneous quantification of the relative or absolute amount of all or selected nucleic acids in the applied nucleic acid sample or mixture. DNA microarrays, therefore, permit an analysis of the expression of multiple (as many as 6800 or more) nucleic acids in parallel (see, e.g., Schena, M. (1996) BioEssays 18(5): 427-431).
  • sequences of the invention may be used to design oligonucleotide primers which are able to amplify defined regions of one or more C. glutamicum genes by a nucleic acid amplification reaction such as the polymerase chain reaction.
  • a nucleic acid amplification reaction such as the polymerase chain reaction.
  • the choice and design of the 5′ or 3′ oligonucleotide primers or of appropriate linkers allows the covalent attachment of the resulting PCR products to the surface of a support medium described above (and also described, for example, Schena, M. et al. (1995) Science 270: 467-470).
  • Nucleic acid microarrays may also be constructed by in situ oligonucleotide synthesis as described by Wodicka, L. et al. (1997) Nature Biotechnology 15: 1359-1367.
  • Photolithographic methods precisely defined regions of the matrix are exposed to light.
  • Protective groups which are photolabile are thereby activated and undergo nucleotide addition, whereas regions that are masked from light do not undergo any modification.
  • Subsequent cycles of protection and light activation permit the synthesis of different oligonucleotides at defined positions.
  • Small, defined regions of the genes of the invention may be synthesized on microarrays by solid phase oligonucleotide synthesis.
  • nucleic acid molecules of the invention present in a sample or mixture of nucleotides may be hybridized to the microarrays.
  • These nucleic acid molecules can be labeled according to standard methods.
  • nucleic acid molecules e.g., mRNA molecules or DNA molecules
  • Hybridization of labeled nucleic acids to microarrays is described (e.g., in Schena, M. et al. (1995) supra; Wodicka, L. et al. (1997), supra; and DeSaizieu A. et al. (1998), supra).
  • Radioactive labels can be detected, for example, as described in Schena, M. et al. (1995) supra) and fluorescent labels may be detected, for example, by the method of Shalon et al. (1996) Genome Research 6: 639-645).
  • sequences of the invention permits comparative analyses of different strains of C. glutamicum or other Corynebacteria .
  • studies of inter-strain variations based on individual transcript profiles and the identification of genes that are important for specific and/or desired strain properties such as pathogenicity, productivity and stress tolerance are facilitated by nucleic acid array methodologies.
  • comparisons of the profile of expression of genes of the invention during the course of a fermentation reaction are possible using nucleic acid array technology.
  • Protein populations of interest include, but are not limited to, the total protein population of C. glutamicum (e.g., in comparison with the protein populations of other organisms), those proteins which are active under specific environmental or metabolic conditions (e.g., during fermentation, at high or low temperature, or at high or low pH), or those proteins which are active during specific phases of growth and development.
  • Protein populations can be analyzed by various well-known techniques, such as gel electrophoresis.
  • Cellular proteins may be obtained, for example, by lysis or extraction, and may be separated from one another using a variety of electrophoretic techniques.
  • Sodium dodecyl sulfate polyacrylamide gel electrophoresis SDS-PAGE
  • Isoelectric focusing polyacrylamide gel electrophoresis Isoelectric focusing polyacrylamide gel electrophoresis (IEF-PAGE) separates proteins by their isoelectric point (which reflects not only the amino acid sequence but also posttranslational modifications of the protein).
  • Another, more preferred method of protein analysis is the consecutive combination of both IEF-PAGE and SDS-PAGE, known as 2-D-gel electrophoresis (described, for example, in Hermann et al. (1998) Electrophoresis 19: 3217-3221; Fountoulakis et al. (1998) Electrophoresis 19: 1193-1202; Langen et al. (1997) Electrophoresis 18: 1184-1192; Antelmann et al. (1997) Electrophoresis 18: 1451-1463).
  • Other separation techniques may also be utilized for protein separation, such as capillary gel electrophoresis; such techniques are well known in the art.
  • Proteins separated by these methodologies can be visualized by standard techniques, such as by staining or labeling. Suitable stains are known in the art, and include Coomassie Brilliant Blue, silver stain, or fluorescent dyes such as Sypro Ruby (Molecular Probes).
  • Suitable stains include Coomassie Brilliant Blue, silver stain, or fluorescent dyes such as Sypro Ruby (Molecular Probes).
  • the inclusion of radioactively labeled amino acids or other protein precursors e.g., 35 S-methionine, 35 S-cysteine, 14 C-labelled amino acids, 15 N-amino acids, 15 NO 3 or 15 NH 4 + or 13 C-labelled amino acids
  • fluorescent labels may be employed. These labeled proteins can be extracted, isolated and separated according to the previously described techniques.
  • Proteins visualized by these techniques can be further analyzed by measuring the amount of dye or label used.
  • the amount of a given protein can be determined quantitatively using, for example, optical methods and can be compared to the amount of other proteins in the same gel or in other gels. Comparisons of proteins on gels can be made, for example, by optical comparison, by spectroscopy, by image scanning and analysis of gels, or through the use of photographic films and screens. Such techniques are well-known in the art.
  • N- and/or C-terminal amino acid sequencing such as Edman degradation
  • mass spectrometry in particular MALDI or ESI techniques (see, e.g., Langen et al. (1997) Electrophoresis 18: 1184-1192)
  • MALDI or ESI techniques see, e.g., Langen et al. (1997) Electrophoresis 18: 1184-1192)
  • the information obtained by these methods can be used to compare patterns of protein presence, activity, or modification between different samples from various biological conditions (e.g., different organisms, time points of fermentation, media conditions, or different biotopes, among others). Data obtained from such experiments alone, or in combination with other techniques, can be used for various applications, such as to compare the behavior of various organisms in a given (e.g., metabolic) situation, to increase the productivity of strains which produce fine chemicals or to increase the efficiency of the production of fine chemicals.
  • various biological conditions e.g., different organisms, time points of fermentation, media conditions, or different biotopes, among others.
  • Data obtained from such experiments alone, or in combination with other techniques can be used for various applications, such as to compare the behavior of various organisms in a given (e.g., metabolic) situation, to increase the productivity of strains which produce fine chemicals or to increase the efficiency of the production of fine chemicals.
  • Corynebacterium glutamicum is equipped with four secondary proline carriers for compatible solutes: Identification, sequencing, and characterization of the proline/ectoine uptake system, ProP, and the ectoine/proline/glycine betaine carrier, EctP,” J. Bacteriol., 180(22): 6005-6012 (1998) AJ004934 dapD Tetrahydrodipicolinate succinylase Wehrmann, A. et al. “Different modes of diaminopimelate synthesis and their (incomplete i ) role in cell wall integrity: A study with Corynebacterium glutamicum ,” J.
  • Gram-positive bacteria with a high DNA G + C content are characterized by a common insertion within their 23S rRNA genes,” J. Gen. Microbiol., 138: 1167-1175 (1992) M85107, 23S rRNA gene insertion sequence Roller, C. et al.
  • Gram-positive bacteria with a high DNA G + C content are M85108 characterized by a common insertion within their 23S rRNA genes
  • Corynebacterium glutamicum aecD gene encodes a C-S uptake carrier; hypothetical protein yhbw lyase with alpha, beta-elimination activity that degrades aminoethylcysteine,” J. Bacteriol., 174(9): 2968-2977 (1992); Tauch, A. et al. “Isoleucine uptake in Corynebacterium glutamicum ATCC 13032 is directed by the brnQ gene product,” Arch. Microbiol., 169(4): 303-312 (1998) S59299 trp Leader gene (promoter) Herry, D. M. et al.
  • a gene encoding arginyl-tRNA synthetase is located in the decarboxylase (partial) upstream region of the lysA gene in Brevibacterium lactofermentum : Regulation of argS-lysA cluster expression by arginine,” J. Bacteriol., 175(22): 7356-7362 (1993) Z21502 dapA; dapB Dihydrodipicolinate synthase; Pisabarro, A. et al.
  • GB_EST5:N23892 434 N23892 yw46f12.s1 Weizmann Olfactory Epithelium Homo sapiens cDNA clone Homo sapiens 38,462 28-DEC-1995 IMAGE: 255311 3′, mRNA sequence.
  • metalB Corynebacterium glutamicum cystathionine gamma-synthase
  • GB_PR3:HSJ659F15 148440 AL096791 Human DNA sequence from clone 659F15 on chromosome Xp11.21-11.4, Homo sapiens 36,190 23-Nov-99 complete sequence.
  • GB_PR2:HS179P9 108260 Z98880 Human DNA sequence from PAC 179P9 on chromosome 6q22.
  • rxa00287 597 GB_IN2:AF144549 7887 AF144549 Aedes albopictus ribosomal protein L34 (rpl34) gene, complete cds. Aedes albopictus 39,828 3-Jun-99 GB_EST15:AA475366 503 AA475366 vh14e09.r1 Soares mouse mammary gland NbMMG Mus musculus cDNA Mus musculus 37,063 18-Jun-97 clone IMAGE: 875464 5′ similar to gb: X87671 M. musculus mRNA for 3BP-1, an SH3 domain binding (MOUSE);, mRNA sequence.
  • MOUSE SH3 domain binding
  • GB_GSS15 AQ641399 569 AQ641399 RPCI93-Dpnll-28C1.
  • TV RPCI93-Dpnll Trypanosoma brucei genomic clone Trypanosoma brucei 39,106 8-Jul-99 RPCI93-Dpnll-28C1, genomic survey sequence.
  • rxa00292 777 GB_PL1:YSCKGD2 2112 M34531 S. cerevisiae dihydrolipoyl transsuccinylase (KGD2) gene, complete cds. Saccharomyces cerevisiae 37,330 27-Apr-93 GB_PL1:SCNUM1 9851 X61236 S.
  • GB_PL2:ATAC007045 68554 AC007045
  • Arabidopsis thaliana chromosome II BAC F23M2 genomic sequence complete Arabidopsis thaliana 43,513 31-MAR-1999 sequence.
  • Bos taurus alpha1-microglobulin/bikunin mRNA complete cds.
  • GB_HTG2:AC006174 203407 AC006174 Home sapiens chromosome 10 clone CIT987SK-1057L21 map 10q25, *** Homo sapiens 38,320 09-DEC-1998 SEQUENCING IN PROGRESS***, 6 unordered pieces.
  • GB_HTG2:AC006174 203407 AC006174 Homo sapiens chromosome 10 clone CIT987SK-1057L21 map 10q25, Homo sapiens 38,320 09-DEC-1998 ***SEQUENCING IN PROGRESS***, 6 unordered pieces.
  • GB_HTG2:AC006174 203407 AC006174 Home sapiens chromosome 10 clone CIT9875K-1057L21 map 10q25, Homo sapiens 37,693 09-DEC-1998 ***SEQUENCING IN PROGRESS***, 6 unordered pieces.
  • tyrocidine synthetase 1 Brevibacillus brevis 40,500 18-Nov-97 (tycA), tyrocidine synthetase 2 (tycB), tyrocidine synthetase 3 (tycC), putative ABC-transporter TycD (tycD), putative ABC-transporter TycE (tycE) and putative thioesterase GrsT homolog (tycF) genes, complete cds.
  • tycA tyrocidine synthetase 1
  • tycB tyrocidine synthetase 2
  • tycC tyrocidine synthetase 3
  • putative ABC-transporter TycD tycD
  • tycE putative ABC-transporter TycE
  • putative thioesterase GrsT homolog tycF
  • rxa00593 471 GB_BA1:MTV025 121125 AL022121 Mycobacterium tuberculosis H37Rv complete genome; segment 155/162.
  • rxa00609 558 GB_HTG3:AC009346 105005 AC009346 Drosophila melanogaster chromosome 3 clone BACR03P13 (D672) RPCI-98 Drosophila melanogaster 31,261 27-Aug-99 03.P.13 map 83A-83B strain y; cn bw sp, ***SEQUENCING IN PROGRESS***, 83 unordered pieces.
  • GB_HTG3:AC009346 105005 AC009346 Drosophila melanogaster chromosome 3 clone BACR03P13 (D672) RPCI-98 Drosophila melanogaster 31,261 27-Aug-99 03.P.13 map 83A-83B strain y; cn bw sp, ***SEQUENCING IN PROGRESS***, 83 unordered pieces.
  • GB_HTG3:AC009346 105005 AC009346 Drosophila melanogaster chromosome 3 clone BACR03P13 (D672) RPCI-98 Drosophila melanogaster 30,072 27-Aug-99 03.P.13 map 83A-83B strain y; cn bw sp, ***SEQUENCING IN PROGRESS***, 83 unordered pieces.
  • GB_PR2 AP000165 100000 AP000165 Homo sapiens genomic DNA, chromosome 21q22.1, D21S226-AML region, Homo sapiens 35,685 20-Nov-99 clone B2344F14-f50E8, segment 1/9, complete sequence.
  • GB_PR3:HS95C20 138849 Z97181 Homo sapiens DNA sequence from PAC 95C20 on chromosome Xp11.3-11.4.
  • Homo sapiens 37,633 23-Nov-99 Contains STSs and the DXS7 locus with GT and GTG repeat polymorphisms, complete sequence.
  • rxa00848 2043 GB_BA1:MTCI65 34331 Z95584 Mycobacterium tuberculosis H37Rv complete genome; segment 50/162.
  • GB_IN1:CET20D3 32679 Z68220 Caenorhabditis elegans cosmid T20D3, complete sequence.
  • GB_BA1 MTCY27 27548 Z95208 Mycobacterium tuberculosis H37Rv complete genome; segment 104/162.
  • Rhodococcus opacus 40,000 24-Sep-97 GB_IN1:CELC14F5 42966 U29082 Caenorhabditis elegans cosmid C14F5.
  • GB_BA2:AF164439 783 AF164439 Mycobacterium smegmatis WhmD (whmD) gene, complete cds; and unknown Mycobacterium smegmatis 57,477 4-Aug-99 gene.
  • GB_BA1:MTV015 1668 AL021840 Mycobacterium tuberculosis H37Rv complete genome; segment 140/162.
  • Danio rerio 36,093 30-Jul-98 GB_STS:G39160 605 G39160 Z13915 Zebrafish AB Danio rerio STS genomic, sequence tagged site.
  • Danio rerio 36,093 30-Jul-98 rxa01763 588 GB_GSS4:AQ701186 454 AQ701186 HS_2129_A2_D04_T7C
  • GB_PR3:HSDJ860P4 156791 AL049594 Human DNA sequence from clone 860P4 on chromosome 20 Contains ESTs, Homo sapiens 35,504 23-Nov-99 STSs, GSSs and a CpG island, complete sequence. rxa01840 654 GB_BA1:D90914 145709 D90914 Synechocystis sp. PCC6803 complete genome, 16/27, 1991550-2137258. Synechocystis sp.
  • GB_HTG4:AC009949 173517 AC009949 Homo sapiens chromosome unknown clone NH0069J07, WORKING DRAFT Homo sapiens 36,544 29-OCT-1999 SEQUENCE, in unordered pieces.
  • GB_HTG4:AC009949 173517 AC009949 Homo sapiens chromosome unknown clone NH0069J07, WORKING DRAFT Homo sapiens 36,544 29-OCT-1999 SEQUENCE, in unordered pieces.
  • NGR234 plasmid pNGR234a section 45 of 46 of the complete Rhizobium sp. NGR234 37,322 12-DEC-1997 plasmid sequence.
  • rxa02127 777 GB_BA1:D90911 143051 D90911
  • Synechocystis sp. PCC6803 complete genome, 13/27, 1576593-1719643.
  • GB_BA2:CGU31230 3005 U31230 Corynebacterium glutamicum Obg protein homolog gene, partial cds, gamma Corynebacterium glutamicum 97,504 2-Aug-96 glutamyl kinase (proB) gene, complete cds, and (unkdh) gene, complete cds.
  • proB glutamyl kinase
  • GB_HTG3:AC009298 165826 AC009298 Homo sapiens clone NH0017106, ***SEQUENCING IN PROGRESS***, 2 Homo sapiens 35,886 13-Aug-99 unordered pieces.
  • glutamicum betP gene Corynebacterium glutamicum 38,346 8-Sep-97 GB_BA1:SHGCPIR 107379 X86780 S. hygroscopicus gene cluster for polyketide immunosuppressant rapamycin.
  • Mycobacterium tuberculosis H37Rv complete genome; segment 25/162.
  • Homo sapiens 36,763 27-Aug-98 rxa02667 717 GB_BA1:MSGY23 40806 AD000016 Mycobacterium tuberculosis sequence from clone y23.
  • GB_PR2:AP000228 75698 AP000228 Homo sapiens genomic DNA, chromosome 21q21.2, LL56-APP region, Homo sapiens 42,616 20-Nov-99 clone: R49K20, complete sequence.
  • rxa02699 2271 GB_GSS12:AQ364540 497 AQ364540 nbxb0061O09r CUGI Rice BAC Library Oryza sativa genomic clone Oryza sativa 37,903 3-Feb-99 nbxb0061O09r, genomic survey sequence.
  • GB_PR4:AC006044 141509 AC006044 Homo sapiens BAC clone NH0539B24 from 7p15.1-p14, complete sequence.
  • GB_PR2:HSAF001552 91526 AF001552
  • Homo sapiens chromosome 16 BAC clone CIT987SK-381E11 complete Homo sapiens 35,352 21-Aug-97 sequence.
  • GB_HTG2:HSDJ139D8 167079 AL096814 Homo sapiens chromosome 6 clone RP1-139D8 map p12.1-21.1, Homo sapiens 36,820 03-DEC-1999 ***SEQUENCING IN PROGRESS***, in unordered pieces.
  • GB_HTG2:HSDJ139D8 167079 AL096814 Homo sapiens chromosome 6 clone RP1-139D8 map p12.1-21.1, Homo sapiens 36,820 03-DEC-1999 ***SEQUENCING IN PROGRESS***, in unordered pieces.
  • GB_IN2 AC004295 84551 AC004295 Drosophila melanogaster DNA sequence (P1 DS08374 (D180)), complete Drosophila melanogaster 40,303 29-Jul-98 sequence.
  • GB_HTG6 AC011647 141830 AC011647 Homo sapiens clone RP11-15D18, ***SEQUENCING IN PROGRESS***, 29 Homo sapiens 38,158 04-DEC-1999 unordered pieces.
  • GB_BA1:MTCY05A6 38631 Z96072 Mycobacterium tuberculosis H37Rv complete genome; segment 120/162.
  • Mycobacterium tuberculosis 61,062 17-Jun-98
  • GB_BA1:CORDTXRAA 2604 M80338 Corynebacterium diphtheriae diphtheria toxin repressor (dtxR) gene, complete Corynebacterium diphtheriae 66,372 26-Apr-93 cds.
  • dtxR Corynebacterium diphtheriae diphtheria toxin repressor
  • GB_HTG3 AC005769 200000 AC005769 Homo sapiens chromosome 4, ***SEQUENCING IN PROGRESS***, 5 Homo sapiens 38,613 21-Aug-99 unordered pieces.

Abstract

Isolated nucleic acid molecules, designated MR nucleic acid molecules, which encode novel MR proteins from Corynebacterium glutamicum are described. The invention also provides antisense nucleic acid molecules, recombinant expression vectors containing MR nucleic acid molecules, and host cells into which the expression vectors have been introduced. The invention still further provides isolated MR proteins, mutated MR proteins, fusion proteins, antigenic peptides and methods for the improvement of production of a desired compound from C. glutamicum based on genetic engineering of MR genes in this organism.

Description

    RELATED APPLICATIONS
  • This application is a divisional application of U.S. patent application Ser. No. 11/006,098, filed Dec. 6, 2004, which is a continuation application of U.S. patent application Ser. No. 09/602,874, filed Jun. 23, 2000, which claims priority to U.S. Provisional Patent Application No. 60/141,031, filed Jun. 25, 1999, U.S. Provisional Patent Application No. 60/142,690, filed Jul. 1, 1999, and also to U.S. Provisional Patent Application No. 60/151,251, filed Aug. 27, 1999. This application also claims priority to German Patent Application No. 19930476.9, filed Jul. 1, 1999, German Patent Application No. 19931419.5, filed Jul. 8, 1999, German Patent Application No. 19931420.9, filed Jul. 8, 1999, German Patent Application No. 19932122.1, filed Jul. 9, 1999, German Patent Application No. 19932128.0, filed Jul. 9, 1999, German Patent Application No. 19932134.5, filed Jul. 9, 1999, German Patent Application No. 19932206.6, filed Jul. 9, 1999, German Patent Application No. 19932207.4, filed Jul. 9, 1999, German Patent Application No. 19933003.4, filed Jul. 14, 1999, German Patent Application No. 19941390.8, filed Aug. 31, 1999, German Patent Application No. 19942088.2, filed Sep. 3, 1999, and German Patent Application No. 19942124.2, filed Sep. 3, 1999. The entire contents of each of the aforementioned applications are hereby expressly incorporated herein by this reference.
  • INCORPORATION OF MATERIAL SUBMITTED ON COMPACT DISCS
  • This application incorporates herein by reference the material contained on the compact discs submitted herewith as part of this application. Specifically, the file “seqlist” (1.06 MB) contained on each of Copy 1, Copy 2 and the CRF copy of the Sequence Listing is hereby incorporated herein by reference. This file was created on Jul. 29, 2006. In addition, the files “Appendix A” (177 KB) and “Appendix B” (61.9 KB) contained on each of the compact disks entitled “Appendices Copy 1” and “Appendices Copy 2” are hereby incorporated herein by reference. Each of these files were created on Jul. 29, 2006.
  • BACKGROUND OF THE INVENTION
  • Certain products and by-products of naturally-occurring metabolic processes in cells have utility in a wide array of industries, including the food, feed, cosmetics, and pharmaceutical industries. These molecules, collectively termed ‘fine chemicals’, include organic acids, both proteinogenic and non-proteinogenic amino acids, nucleotides and nucleosides, lipids and fatty acids, diols, carbohydrates, aromatic compounds, vitamins and cofactors, and enzymes. Their production is most conveniently performed through the large-scale culture of bacteria developed to produce and secrete large quantities of one or more desired molecules. One particularly useful organism for this purpose is Corynebacterium glutamicum, a gram positive, nonpathogenic bacterium. Through strain selection, a number of mutant strains have been developed which produce an array of desirable compounds. However, selection of strains improved for the production of a particular molecule is a time-consuming and difficult process.
  • SUMMARY OF THE INVENTION
  • The invention provides novel bacterial nucleic acid molecules which have a variety of uses. These uses include the identification of microorganisms which can be used to produce fine chemicals, the modulation of fine chemical production in C. glutamicum or related bacteria, the typing or identification of C. glutamicum or related bacteria, as reference points for mapping the C. glutamicum genome, and as markers for transformation. These novel nucleic acid molecules encode proteins, referred to herein as metabolic regulatory (MR) proteins.
  • C. glutamicum is a gram positive, aerobic bacterium which is commonly used in industry for the large-scale production of a variety of fine chemicals, and also for the degradation of hydrocarbons (such as in petroleum spills) and for the oxidation of terpenoids. The MR nucleic acid molecules of the invention, therefore, can be used to identify microorganisms which can be used to produce fine chemicals, e.g., by fermentation processes. Modulation of the expression of the MR nucleic acids of the invention, or modification of the sequence of the MR nucleic acid molecules of the invention, can be used to modulate the production of one or more fine chemicals from a microorganism (e.g., to improve the yield or production of one or more fine chemicals from a Corynebacterium or Brevibacterium species).
  • The MR nucleic acids of the invention may also be used to identify an organism as being Corynebacterium glutamicum or a close relative thereof, or to identify the presence of C. glutamicum or a relative thereof in a mixed population of microorganisms. The invention provides the nucleic acid sequences of a number of C. glutamicum genes; by probing the extracted genomic DNA of a culture of a unique or mixed population of microorganisms under stringent conditions with a probe spanning a region of a C. glutamicum gene which is unique to this organism, one can ascertain whether this organism is present. Although Corynebacterium glutamicum itself is nonpathogenic, it is related to species pathogenic in humans, such as Corynebacterium diphtheriae (the causative agent of diphtheria); the detection of such organisms is of significant clinical relevance.
  • The MR nucleic acid molecules of the invention may also serve as reference points for mapping of the C. glutamicum genome, or of genomes of related organisms. Similarly, these molecules, or variants or portions thereof, may serve as markers for genetically engineered Corynebacterium or Brevibacterium species.
  • e.g. The MR proteins encoded by the novel nucleic acid molecules of the invention are capable of, for example, performing a function involved in the transcriptional, translational, or posttranslational regulation of proteins important for the normal metabolic functioning of cells. Given the availability of cloning vectors for use in Corynebacterium glutamicum, such as those disclosed in Sinskey et al., U.S. Pat. No. 4,649,119, and techniques for genetic manipulation of C. glutamicum and the related Brevibacterium species (e.g., lactofermentum) (Yoshihama et al, J. Bacteriol. 162: 591-597 (1985); Katsumata et al., J. Bacteriol. 159: 306-311 (1984); and Santamaria et al., J. Gen. Microbiol. 130: 2237-2246 (1984)), the nucleic acid molecules of the invention may be utilized in the genetic engineering of this organism to make it a better or more efficient producer of one or more fine chemicals.
  • This improved yield, production and/or efficiency of production of a fine chemical may be due to a direct effect of manipulation of a gene of the invention, or it may be due to an indirect effect of such manipulation. Specifically, alterations in C. glutamicum MR proteins which normally regulate the yield, production and/or efficiency of production of a fine chemical metabolic pathways may have a direct impact on the overall production or rate of production of one or more of these desired compounds from this organism. Alterations in the proteins involved in these metabolic pathways may also have an indirect impact on the yield, production and/or efficiency of production of a desired fine chemical. Regulation of metabolism is necessarily complex, and the regulatory mechanisms governing different pathways may intersect at multiple points such that more than one pathway can be rapidly adjusted in accordance with a particular cellular event. This enables the modification of a regulatory protein for one pathway to have an impact on the regulation of many other pathways as well, some of which may be involved in the biosynthesis or degradation of a desired fine chemical. In this indirect fashion, the modulation of action of an MR protein may have an impact on the production of a fine chemical produced by a pathway different from one which that MR protein directly regulates.
  • The nucleic acid and protein molecules of the invention may be utilized to directly improve the yield, production, and/or efficiency of production of one or more desired fine chemicals from Corynebacterium glutamicum. Using recombinant genetic techniques well known in the art, one or more of the regulatory proteins of the invention may be manipulated such that its function is modulated. For example, the mutation of an MR protein involved in the repression of transcription of a gene encoding an enzyme which is required for the biosynthesis of an amino acid such that it no longer is able to repress transcription may result in an increase in production of that amino acid. Similarly, the alteration of activity of an MR protein resulting in increased translation or activating posttranslational modification of a C. glutamicum protein involved in the biosynthesis of a desired fine chemical may in turn increase the production of that chemical. The opposite situation may also be of benefit: by increasing the repression of transcription or translation, or by posttranslational negative modification of a C. glutamicum protein involved in the regulation of a degradative pathway for a compound, one may increase the production of this chemical. In each case, the overall yield or rate of production of the desired fine chemical may be increased.
  • It is also possible that such alterations in the protein and nucleotide molecules of the invention may improve the yield, production, and/or efficiency of production of fine chemicals through indirect mechanisms. The metabolism of any one compound is necessarily intertwined with other biosynthetic and degradative pathways within the cell, and necessary cofactors, intermediates, or substrates in one pathway are likely supplied or limited by another such pathway. Therefore, by modulating the activity of one or more of the regulatory proteins of the invention, the production or efficiency of activity of another fine chemical biosynthetic or degradative pathway may be impacted. Further, the manipulation of one or more regulatory proteins may increase the overall ability of the cell to grow and multiply in culture, particularly in large-scale fermentative culture, where growth conditions may be suboptimal. For example, by mutating an MR protein of the invention which would normally cause a repression in the biosynthesis of nucleotides in response to suboptimal extracellular supplies of nutrients (thereby preventing cell division) such that it is decreased in repressor ability, one may increase the biosynthesis of nucleotides and perhaps increase cell division. Changes in MR proteins which result in increased cell growth and division in culture may result in an increase in yield, production, and/or efficiency of production of one or more desired fine chemicals from the culture, due at least to the increased number of cells producing the chemical in the culture.
  • The invention provides novel nucleic acid molecules which encode proteins, referred to herein as metabolic pathway proteins (MR), which are capable of, for example, performing an enzymatic step involved in the transcriptional, translational, or posttranslational regulation of metabolic pathways in C. glutamicum. Nucleic acid molecules encoding an MR protein are referred to herein as MR nucleic acid molecules. In a preferred embodiment, the MR protein participates in the transcriptional, translational, or posttranslational regulation of one or more metabolic pathways. Examples of such proteins include those encoded by the genes set forth in Table 1.
  • Accordingly, one aspect of the invention pertains to isolated nucleic acid molecules (e.g., cDNAs, DNAs, or RNAs) comprising a nucleotide sequence encoding an MR protein or biologically active portions thereof, as well as nucleic acid fragments suitable as primers or hybridization probes for the detection or amplification of MR-encoding nucleic acid (e.g., DNA or mRNA). In particularly preferred embodiments, the isolated nucleic acid molecule comprises one of the nucleotide sequences set forth in Appendix A or the coding region or a complement thereof of one of these nucleotide sequences. In other particularly preferred embodiments, the isolated nucleic acid molecule of the invention comprises a nucleotide sequence which hybridizes to or is at least about 50%, preferably at least about 60%, more preferably at least about 70%, 80% or 90%, and even more preferably at least about 95%, 96%, 97%, 98%, 99% or more homologous to a nucleotide sequence set forth in Appendix A, or a portion thereof. In other preferred embodiments, the isolated nucleic acid molecule encodes one of the amino acid sequences set forth in Appendix B. The preferred MR proteins of the present invention also preferably possess at least one of the MR activities described herein.
  • In another embodiment, the isolated nucleic acid molecule encodes a protein or portion thereof wherein the protein or portion thereof includes an amino acid sequence which is sufficiently homologous to an amino acid sequence of Appendix B, e.g., sufficiently homologous to an amino acid sequence of Appendix B such that the protein or portion thereof maintains an MR activity. Preferably, the protein or portion thereof encoded by the nucleic acid molecule maintains the ability to transcriptionally, translationally, or posttranslationally regulate a metabolic pathway in C. glutamicum. In one embodiment, the protein encoded by the nucleic acid molecule is at least about 50%, preferably at least about 60%, and more preferably at least about 70%, 80%, or 90% and most preferably at least about 95%, 96%, 97%, 98%, or 99% or more homologous to an amino acid sequence of Appendix B (e.g., an entire amino acid sequence selected from those sequences set forth in Appendix B). In another preferred embodiment, the protein is a full length C. glutamicum protein which is substantially homologous to an entire amino acid sequence of Appendix B (encoded by an open reading frame shown in Appendix A).
  • In another preferred embodiment, the isolated nucleic acid molecule is derived from C. glutamicum and encodes a protein (e.g., an MR fusion protein) which includes a biologically active domain which is at least about 50% or more homologous to one of the amino acid sequences of Appendix B and is able to transcriptionally, translationally, or posttranslationally regulate a metabolic pathway in C. glutamicum, or has one or more of the activities set forth in Table 1, and which also includes heterologous nucleic acid sequences encoding a heterologous polypeptide or regulatory regions.
  • In another embodiment, the isolated nucleic acid molecule is at least 15 nucleotides in length and hybridizes under stringent conditions to a nucleic acid molecule comprising a nucleotide sequence of Appendix A. Preferably, the isolated nucleic acid molecule corresponds to a naturally-occurring nucleic acid molecule. More preferably, the isolated nucleic acid encodes a naturally-occurring C. glutamicum MR protein, or a biologically active portion thereof.
  • Another aspect of the invention pertains to vectors, e.g., recombinant expression vectors, containing the nucleic acid molecules of the invention, and host cells into which such vectors have been introduced. In one embodiment, such a host cell is used to produce an MR protein by culturing the host cell in a suitable medium. The MR protein can be then isolated from the medium or the host cell.
  • Yet another aspect of the invention pertains to a genetically altered microorganism in which an MR gene has been introduced or altered. In one embodiment, the genome of the microorganism has been altered by introduction of a nucleic acid molecule of the invention encoding wild-type or mutated MR sequence as a transgene. In another embodiment, an endogenous MR gene within the genome of the microorganism has been altered, e.g., functionally disrupted, by homologous recombination with an altered MR gene. In another embodiment, an endogenous or introduced MR gene in a microorganism has been altered by one or more point mutations, deletions, or inversions, but still encodes a functional MR protein. In still another embodiment, one or more of the regulatory regions (e.g., a promoter, repressor, or inducer) of an MR gene in a microorganism has been altered (e.g., by deletion, truncation, inversion, or point mutation) such that the expression of the MR gene is modulated. In a preferred embodiment, the microorganism belongs to the genus Corynebacterium or Brevibacterium, with Corynebacterium glutamicum being particularly preferred. In a preferred embodiment, the microorganism is also utilized for the production of a desired compound, such as an amino acid, with lysine being particularly preferred.
  • In another aspect, the invention provides a method of identifying the presence or activity of Cornyebacterium diphtheriae in a subject. This method includes detection of one or more of the nucleic acid or amino acid sequences of the invention (e.g., the sequences set forth in Appendix A or Appendix B) in a subject, thereby detecting the presence or activity of Corynebacterium diphtheriae in the subject. Still another aspect of the invention pertains to an isolated MR protein or a portion, e.g., a biologically active portion, thereof. In a preferred embodiment, the isolated MR protein or portion thereof transcriptionally, translationally, or posttranslationally regulates one or more metabolic pathways in C. glutamicum. In another preferred embodiment, the isolated MR protein or portion thereof is sufficiently homologous to an amino acid sequence of Appendix B such that the protein or portion thereof maintains the ability to transcriptionally, translationally, or posttranslationally regulate one or more metabolic pathways in C. glutamicum.
  • The invention also provides an isolated preparation of an MR protein. In preferred embodiments, the MR protein comprises an amino acid sequence of Appendix B. In another preferred embodiment, the invention pertains to an isolated full length protein which is substantially homologous to an entire amino acid sequence of Appendix B (encoded by an open reading frame set forth in Appendix A). In yet another embodiment, the protein is at least about 50%, preferably at least about 60%, and more preferably at least about 70%, 80%, or 90%, and most preferably at least about 95%, 96%, 97%, 98%, or 99% or more homologous to an entire amino acid sequence of Appendix B. In other embodiments, the isolated MR protein comprises an amino acid sequence which is at least about 50% or more homologous to one of the amino acid sequences of Appendix B and is able to transcriptionally, translationally, or posttranslationally regulate one or more metabolic pathways in C. glutamicum, or has one or more of the activities set forth in Table 1.
  • Alternatively, the isolated MR protein can comprise an amino acid sequence which is encoded by a nucleotide sequence which hybridizes, e.g., hybridizes under stringent conditions, or is at least about 50%, preferably at least about 60%, more preferably at least about 70%, 80%, or 90%, and even more preferably at least about 95%, 96%, 97%, 98,%, or 99% or more homologous, to a nucleotide sequence of Appendix B. It is also preferred that the preferred forms of MR proteins also have one or more of the MR bioactivities described herein.
  • The MR polypeptide, or a biologically active portion thereof, can be operatively linked to a non-MR polypeptide to form a fusion protein. In preferred embodiments, this fusion protein has an activity which differs from that of the MR protein alone. In other preferred embodiments, this fusion protein transcriptionally, translationally, or posttranslationally regulates one or more metabolic pathways in C. glutamicum. In particularly preferred embodiments, integration of this fusion protein into a host cell modulates production of a desired compound from the cell. In another aspect, the invention provides methods for screening molecules which modulate the activity of an MR protein, either by interacting with the protein itself or a substrate or binding partner of the MR protein, or by modulating the transcription or translation of an MR nucleic acid molecule of the invention. Another aspect of the invention pertains to a method for producing a fine chemical. This method involves the culturing of a cell containing a vector directing the expression of an MR nucleic acid molecule of the invention, such that a fine chemical is produced. In a preferred embodiment, this method further includes the step of obtaining a cell containing such a vector, in which a cell is transfected with a vector directing the expression of an MR nucleic acid. In another preferred embodiment, this method further includes the step of recovering the fine chemical from the culture. In a particularly preferred embodiment, the cell is from the genus Corynebacterium or Brevibacterium, or is selected from those strains set forth in Table 3.
  • Another aspect of the invention pertains to methods for modulating production of a molecule from a microorganism. Such methods include contacting the cell with an agent which modulates MR protein activity or MR nucleic acid expression such that a cell associated activity is altered relative to this same activity in the absence of the agent. In a preferred embodiment, the cell is modulated for one or more C. glutamicum metabolic pathway regulatory systems, such that the yields or rate of production of a desired fine chemical by this microorganism is improved. The agent which modulates MR protein activity can be an agent which stimulates MR protein activity or MR nucleic acid expression. Examples of agents which stimulate MR protein activity or MR nucleic acid expression include small molecules, active MR proteins, and nucleic acids encoding MR proteins that have been introduced into the cell. Examples of agents which inhibit MR activity or expression include small molecules and antisense MR nucleic acid molecules.
  • Another aspect of the invention pertains to methods for modulating yields of a desired compound from a cell, involving the introduction of a wild-type or mutant MR gene into a cell, either maintained on a separate plasmid or integrated into the genome of the host cell. If integrated into the genome, such integration can be random, or it can take place by homologous recombination such that the native gene is replaced by the introduced copy, causing the production of the desired compound from the cell to be modulated. In a preferred embodiment, said yields are increased. In another preferred embodiment, said chemical is a fine chemical. In a particularly preferred embodiment, said fine chemical is an amino acid. In especially preferred embodiments, said amino acid is L-lysine.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention provides MR nucleic acid and protein molecules which are involved in the regulation of metabolism in Corynebacterium glutamicum, including regulation of fine chemical metabolism. The molecules of the invention may be utilized in the modulation of production of fine chemicals from microorganisms, such as C. glutamicum, either directly (e.g., where modulation of the activity of a lysine biosynthesis regulatory protein has a direct impact on the yield, production, and/or efficiency of production of lysine from that organism), or may have an indirect impact which nonetheless results in an increase in yield, production, and/or efficiency of production of the desired compound (e.g., where modulation of the regulation of a nucleotide biosynthesis protein has an impact on the production of an organic acid or a fatty acid from the bacterium, perhaps due to concomitant regulatory alterations in the biosynthetic or degradation pathways for these chemicals in response to the altered regulation of nucleotide biosynthesis). Aspects of the invention are further explicated below.
  • I. Fine Chemicals
  • The term ‘fine chemical’ is art-recognized and includes molecules produced by an organism which have applications in various industries, such as, but not limited to, the pharmaceutical, agriculture, and cosmetics industries. Such compounds include organic acids, such as tartaric acid, itaconic acid, and diaminopimelic acid, both proteinogenic and non-proteinogenic amino acids, purine and pyrimidine bases, nucleosides, and nucleotides (as described e.g. in Kuninaka, A. (1996) Nucleotides and related compounds, p. 561-612, in Biotechnology vol. 6, Rehm et al., eds. VCH: Weinheim, and references contained therein), lipids, both saturated and unsaturated fatty acids (e.g., arachidonic acid), diols (e.g., propane diol, and butane diol), carbohydrates (e.g., hyaluronic acid and trehalose), aromatic compounds (e.g., aromatic amines, vanillin, and indigo), vitamins and cofactors (as described in Ullmann's Encyclopedia of Industrial Chemistry, vol. A27, “Vitamins”, p. 443-613 (1996) VCH: Weinheim and references therein; and Ong, A. S., Niki, E. & Packer, L. (1995) “Nutrition, Lipids, Health, and Disease” Proceedings of the UNESCO/Confederation of Scientific and Technological Associations in Malaysia, and the Society for Free Radical Research—Asia, held Sep. 1-3, 1994 at Penang, Malaysia, AOCS Press, (1995)), enzymes, polyketides (Cane et al. (1998) Science 282: 63-68), and all other chemicals described in Gutcho (1983) Chemicals by Fermentation, Noyes Data Corporation, ISBN: 0818805086 and references therein. The metabolism and uses of certain of these fine chemicals are further explicated below.
  • A. Amino Acid Metabolism and Uses
  • Amino acids comprise the basic structural units of all proteins, and as such are essential for normal cellular functioning in all organisms. The term “amino acid” is art-recognized. The proteinogenic amino acids, of which there are 20 species, serve as structural units for proteins, in which they are linked by peptide bonds, while the nonproteinogenic amino acids (hundreds of which are known) are not normally found in proteins (see Ulmann's Encyclopedia of Industrial Chemistry, vol. A2, p. 57-97 VCH: Weinheim (1985)). Amino acids may be in the D- or L-optical configuration, though L-amino acids are generally the only type found in naturally-occurring proteins. Biosynthetic and degradative pathways of each of the 20 proteinogenic amino acids have been well characterized in both prokaryotic and eukaryotic cells (see, for example, Stryer, L. Biochemistry, 3rd edition, pages 578-590 (1988)). The ‘essential’ amino acids (histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, and valine), so named because they are generally a nutritional requirement due to the complexity of their biosyntheses, are readily converted by simple biosynthetic pathways to the remaining 11 ‘nonessential’ amino acids (alanine, arginine, asparagine, aspartate, cysteine, glutamate, glutamine, glycine, proline, serine, and tyrosine). Higher animals do retain the ability to synthesize some of these amino acids, but the essential amino acids must be supplied from the diet in order for normal protein synthesis to occur.
  • Aside from their function in protein biosynthesis, these amino acids are interesting chemicals in their own right, and many have been found to have various applications in the food, feed, chemical, cosmetics, agriculture, and pharmaceutical industries. Lysine is an important amino acid in the nutrition not only of humans, but also of monogastric animals such as poultry and swine. Glutamate is most commonly used as a flavor additive (mono-sodium glutamate, MSG) and is widely used throughout the food industry, as are aspartate, phenylalanine, glycine, and cysteine. Glycine, L-methionine and tryptophan are all utilized in the pharmaceutical industry. Glutamine, valine, leucine, isoleucine, histidine, arginine, proline, serine and alanine are of use in both the pharmaceutical and cosmetics industries. Threonine, tryptophan, and D/L-methionine are common feed additives. (Leuchtenberger, W. (1996) Amino aids—technical production and use, p. 466-502 in Rehm et al. (eds.) Biotechnology vol. 6, chapter 14a, VCH: Weinheim). Additionally, these amino acids have been found to be useful as precursors for the synthesis of synthetic amino acids and proteins, such as N-acetylcysteine, S-carboxymethyl-L-cysteine, (S)-5-hydroxytryptophan, and others described in Ulmann's Encyclopedia of Industrial Chemistry, vol. A2, p. 57-97, VCH: Weinheim, 1985.
  • The biosynthesis of these natural amino acids in organisms capable of producing them, such as bacteria, has been well characterized (for review of bacterial amino acid biosynthesis and regulation thereof, see Umbarger, H. E. (1978) Ann. Rev. Biochem. 47: 533-606). Glutamate is synthesized by the reductive amination of α-ketoglutarate, an intermediate in the citric acid cycle. Glutamine, proline, and arginine are each subsequently produced from glutamate. The biosynthesis of serine is a three-step process beginning with 3-phosphoglycerate (an intermediate in glycolysis), and resulting in this amino acid after oxidation, transamination, and hydrolysis steps. Both cysteine and glycine are produced from serine; the former by the condensation of homocysteine with serine, and the latter by the transferal of the side-chain β-carbon atom to tetrahydrofolate, in a reaction catalyzed by serine transhydroxymethylase. Phenylalanine, and tyrosine are synthesized from the glycolytic and pentose phosphate pathway precursors erythrose 4-phosphate and phosphoenolpyruvate in a 9-step biosynthetic pathway that differ only at the final two steps after synthesis of prephenate. Tryptophan is also produced from these two initial molecules, but its synthesis is an 11-step pathway. Tyrosine may also be synthesized from phenylalanine, in a reaction catalyzed by phenylalanine hydroxylase. Alanine, valine, and leucine are all biosynthetic products of pyruvate, the final product of glycolysis. Aspartate is formed from oxaloacetate, an intermediate of the citric acid cycle. Asparagine, methionine, threonine, and lysine are each produced by the conversion of aspartate. Isoleucine is formed from threonine. A complex 9-step pathway results in the production of histidine from 5-phosphoribosyl-1-pyrophosphate, an activated sugar.
  • Amino acids in excess of the protein synthesis needs of the cell cannot be stored, and are instead degraded to provide intermediates for the major metabolic pathways of the cell (for review see Stryer, L. Biochemistry 3rd ed. Ch. 21 “Amino Acid Degradation and the Urea Cycle” p. 495-516 (1988)). Although the cell is able to convert unwanted amino acids into useful metabolic intermediates, amino acid production is costly in terms of energy, precursor molecules, and the enzymes necessary to synthesize them. Thus it is not surprising that amino acid biosynthesis is regulated by feedback inhibition, in which the presence of a particular amino acid serves to slow or entirely stop its own production (for overview of feedback mechanisms in amino acid biosynthetic pathways, see Stryer, L. Biochemistry, 3rd ed. Ch. 24: “Biosynthesis of Amino Acids and Heme” p. 575-600 (1988)). Thus, the output of any particular amino acid is limited by the amount of that amino acid present in the cell.
  • B. Vitamin, Cofactor, and Nutraceutical Metabolism and Uses
  • Vitamins, cofactors, and nutraceuticals comprise another group of molecules which the higher animals have lost the ability to synthesize and so must ingest, although they are readily synthesized by other organisms such as bacteria. These molecules are either bioactive substances themselves, or are precursors of biologically active substances which may serve as electron carriers or intermediates in a variety of metabolic pathways. Aside from their nutritive value, these compounds also have significant industrial value as coloring agents, antioxidants, and catalysts or other processing aids. (For an overview of the structure, activity, and industrial applications of these compounds, see, for example, Ullman's Encyclopedia of Industrial Chemistry, “Vitamins” vol. A27, p. 443-613, VCH: Weinheim, 1996.) The term “vitamin” is art-recognized, and includes nutrients which are required by an organism for normal functioning, but which that organism cannot synthesize by itself. The group of vitamins may encompass cofactors and nutraceutical compounds. The language “cofactor” includes nonproteinaceous compounds required for a normal enzymatic activity to occur. Such compounds may be organic or inorganic; the cofactor molecules of the invention are preferably organic. The term “nutraceutical” includes dietary supplements having health benefits in plants and animals, particularly humans. Examples of such molecules are vitamins, antioxidants, and also certain lipids (e.g., polyunsaturated fatty acids).
  • The biosynthesis of these molecules in organisms capable of producing them, such as bacteria, has been largely characterized (Ullman's Encyclopedia of Industrial Chemistry, “Vitamins” vol. A27, p. 443-613, VCH: Weinheim, 1996; Michal, G. (1999) Biochemical Pathways: An Atlas of Biochemistry and Molecular Biology, John Wiley & Sons; Ong, A. S., Niki, E. & Packer, L. (1995) “Nutrition, Lipids, Health, and Disease” Proceedings of the UNESCO/Confederation of Scientific and Technological Associations in Malaysia, and the Society for Free Radical Research—Asia, held Sep. 1-3, 1994 at Penang, Malaysia, AOCS Press: Champaign, Ill. X, 374 S).
  • Thiamin (vitamin B1) is produced by the chemical coupling of pyrimidine and thiazole moieties. Riboflavin (vitamin B2) is synthesized from guanosine-5′-triphosphate (GTP) and ribose-5′-phosphate. Riboflavin, in turn, is utilized for the synthesis of flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD). The family of compounds collectively termed ‘vitamin B6’ (e.g., pyridoxine, pyridoxamine, pyridoxa-5′-phosphate, and the commercially used pyridoxin hydrochloride) are all derivatives of the common structural unit, 5-hydroxy-6-methylpyridine. Pantothenate (pantothenic acid, (R)-(+)-N-(2,4-dihydroxy-3,3-dimethyl-1-oxobutyl)-β-alanine) can be produced either by chemical synthesis or by fermentation. The final steps in pantothenate biosynthesis consist of the ATP-driven condensation of β-alanine and pantoic acid. The enzymes responsible for the biosynthesis steps for the conversion to pantoic acid, to β-alanine and for the condensation to panthotenic acid are known. The metabolically active form of pantothenate is Coenzyme A, for which the biosynthesis proceeds in 5 enzymatic steps. Pantothenate, pyridoxal-5′-phosphate, cysteine and ATP are the precursors of Coenzyme A. These enzymes not only catalyze the formation of panthothante, but also the production of (R)-pantoic acid, (R)-pantolacton, (R)-panthenol (provitamin B5), pantetheine (and its derivatives) and coenzyme A.
  • Biotin biosynthesis from the precursor molecule pimeloyl-CoA in microorganisms has been studied in detail and several of the genes involved have been identified. Many of the corresponding proteins have been found to also be involved in Fe-cluster synthesis and are members of the nifS class of proteins. Lipoic acid is derived from octanoic acid, and serves as a coenzyme in energy metabolism, where it becomes part of the pyruvate dehydrogenase complex and the α-ketoglutarate dehydrogenase complex. The folates are a group of substances which are all derivatives of folic acid, which is turn is derived from L-glutamic acid, p-amino-benzoic acid and 6-methylpterin. The biosynthesis of folic acid and its derivatives, starting from the metabolism intermediates guanosine-5′-triphosphate (GTP), L-glutamic acid and p-amino-benzoic acid has been studied in detail in certain microorganisms.
  • Corrinoids (such as the cobalamines and particularly vitamin B12) and porphyrines belong to a group of chemicals characterized by a tetrapyrole ring system. The biosynthesis of vitamin B12 is sufficiently complex that it has not yet been completely characterized, but many of the enzymes and substrates involved are now known. Nicotinic acid (nicotinate), and nicotinamide are pyridine derivatives which are also termed ‘niacin’. Niacin is the precursor of the important coenzymes NAD (nicotinamide adenine dinucleotide) and NADP (nicotinamide adenine dinucleotide phosphate) and their reduced forms.
  • The large-scale production of these compounds has largely relied on cell-free chemical syntheses, though some of these chemicals have also been produced by large-scale culture of microorganisms, such as riboflavin, Vitamin B6, pantothenate, and biotin. Only Vitamin B12 is produced solely by fermentation, due to the complexity of its synthesis. In vitro methodologies require significant inputs of materials and time, often at great cost.
  • C. Purine, Pyrimidine, Nucleoside and Nucleotide Metabolism and Uses
  • Purine and pyrimidine metabolism genes and their corresponding proteins are important targets for the therapy of tumor diseases and viral infections. The language “purine” or “pyrimidine” includes the nitrogenous bases which are constituents of nucleic acids, co-enzymes, and nucleotides. The term “nucleotide” includes the basic structural units of nucleic acid molecules, which are comprised of a nitrogenous base, a pentose sugar (in the case of RNA, the sugar is ribose; in the case of DNA, the sugar is D-deoxyribose), and phosphoric acid. The language “nucleoside” includes molecules which serve as precursors to nucleotides, but which are lacking the phosphoric acid moiety that nucleotides possess. By inhibiting the biosynthesis of these molecules, or their mobilization to form nucleic acid molecules, it is possible to inhibit RNA and DNA synthesis; by inhibiting this activity in a fashion targeted to cancerous cells, the ability of tumor cells to divide and replicate may be inhibited. Additionally, there are nucleotides which do not form nucleic acid molecules, but rather serve as energy stores (i.e., AMP) or as coenzymes (i.e., FAD and NAD).
  • Several publications have described the use of these chemicals for these medical indications, by influencing purine and/or pyrimidine metabolism (e.g. Christopherson, R. I. and Lyons, S. D. (1990) “Potent inhibitors of de novo pyrimidine and purine biosynthesis as chemotherapeutic agents.” Med. Res. Reviews 10: 505-548). Studies of enzymes involved in purine and pyrimidine metabolism have been focused on the development of new drugs which can be used, for example, as immunosuppressants or anti-proliferants (Smith, J. L., (1995) “Enzymes in nucleotide synthesis.” Curr. Opin. Struct. Biol. 5: 752-757; (1995) Biochem Soc. Transact. 23: 877-902). However, purine and pyrimidine bases, nucleosides and nucleotides have other utilities: as intermediates in the biosynthesis of several fine chemicals (e.g., thiamine, S-adenosyl-methionine, folates, or riboflavin), as energy carriers for the cell (e.g., ATP or GTP), and for chemicals themselves, commonly used as flavor enhancers (e.g., IMP or GMP) or for several medicinal applications (see, for example, Kuninaka, A. (1996) Nucleotides and Related Compounds in Biotechnology vol. 6, Rehm et al., eds. VCH: Weinheim, p. 561-612). Also, enzymes involved in purine, pyrimidine, nucleoside, or nucleotide metabolism are increasingly serving as targets against which chemicals for crop protection, including fungicides, herbicides and insecticides, are developed.
  • The metabolism of these compounds in bacteria has been characterized (for reviews see, for example, Zalkin, H. and Dixon, J. E. (1992) “de novo purine nucleotide biosynthesis”, in: Progress in Nucleic Acid Research and Molecular Biology, vol. 42, Academic Press: p. 259-287; and Michal, G. (1999) “Nucleotides and Nucleosides”, Chapter 8 in: Biochemical Pathways: An Atlas of Biochemistry and Molecular Biology, Wiley: New York). Purine metabolism has been the subject of intensive research, and is essential to the normal functioning of the cell. Impaired purine metabolism in higher animals can cause severe disease, such as gout. Purine nucleotides are synthesized from ribose-5-phosphate, in a series of steps through the intermediate compound inosine-5′-phosphate (IMP), resulting in the production of guanosine-5′-monophosphate (GMP) or adenosine-5′-monophosphate (AMP), from which the triphosphate forms utilized as nucleotides are readily formed. These compounds are also utilized as energy stores, so their degradation provides energy for many different biochemical processes in the cell. Pyrimidine biosynthesis proceeds by the formation of uridine-5′-monophosphate (UMP) from ribose-5-phosphate. UMP, in turn, is converted to cytidine-5′-triphosphate (CTP). The deoxy-forms of all of these nucleotides are produced in a one step reduction reaction from the diphosphate ribose form of the nucleotide to the diphosphate deoxyribose form of the nucleotide. Upon phosphorylation, these molecules are able to participate in DNA synthesis.
  • D. Trehalose Metabolism and Uses
  • Trehalose consists of two glucose molecules, bound in α,α-1,1 linkage. It is commonly used in the food industry as a sweetener, an additive for dried or frozen foods, and in beverages. However, it also has applications in the pharmaceutical, cosmetics and biotechnology industries (see, for example, Nishimoto et al., (1998) U.S. Pat. No. 5,759,610; Singer, M. A. and Lindquist, S. (1998) Trends Biotech. 16: 460-467; Paiva, C. L. A. and Panek, A. D. (1996) Biotech. Ann. Rev. 2: 293-314; and Shiosaka, M. (1997) J. Japan 172: 97-102). Trehalose is produced by enzymes from many microorganisms and is naturally released into the surrounding medium, from which it can be collected using methods known in the art.
  • II. Mechanisms of Metabolic Regulation
  • All living cells have complex catabolic and anabolic metabolic capabilities with many interconnected pathways. In order to maintain a balance between the various parts of this extremely complex metabolic network, the cell employs a finely-tuned regulatory network. By regulating enzyme synthesis and enzyme activity, either independently or simultaneously, the cell is able to control the activity of disparate metabolic pathways to reflect the changing needs of the cell.
  • The induction or repression of enzyme synthesis may occur at either the level of transcription or translation, or both. Gene expression in prokaryotes is regulated by several mechanisms at the level of transcription (for review see e.g., Lewin, B (1990) Genes IV, Part 3: “Controlling prokaryotic genes by transcription”, Oxford University Press: Oxford, p. 213-301, and references therein, and Michal, G. (1999) Biochemical Pathways: An Atlas of Biochemistry and Molecular Biology, John Wiley & Sons). All such known regulatory processes are mediated by additional genes, which themselves respond to external influences of various kinds (e.g., temperature, nutrient availability, or light). Exemplary protein factors which have been implicated in this type of regulation include the transcription factors. These are proteins which bind to DNA, thereby either increasing the expression of a gene (positive regulation, as in the case of e.g. the ara operon from E. coli) or decreasing gene expression (negative regulation, as in the case of the lac operon from E. coli). These expression-modulating transcription factors can themselves be the subject of regulation. Their activity can, for example, be regulated by the binding of low molecular weight compounds to the DNA-binding protein, thereby stimulating (as in the case of arabinose for the ara operon) or inhibiting (as in the case of the lactose for the lac operon) the binding of these proteins to the appropriate binding site on the DNA (see, for example, Helmann, J. D. and Chamberlin, M. J. (1988) “Structure and function of bacterial sigma factors.” Ann. Rev. Biochem. 57: 839-872; Adhya, S. (1995) “The lac and gal operons today” and Boos, W. et al., “The maltose system.”, both in: Regulation of Gene Expression in Escherichia coli (Lin, E. C. C. and Lynch, A. S., eds.) Chapman & Hall: New York, p. 181-200 and 201-229; and Moran, C. P. (1993) “RNA polymerase and transcription factors.” in: Bacillus subtilis and other gram-positive bacteria, Sonenshein, A. L. et al., eds. ASM: Washington, D.C., p. 653-667.)
  • Aside from the transcriptional level, protein synthesis is also often regulated at the level of translation. There are multiple mechanisms by which such regulation may occur, including alteration of the ability of the ribosome to bind to one or more mRNAs, binding of the ribosome to the mRNA, the maintenance or removal of mRNA secondary structure, the utilization of common or less common codons for a particular gene, the degree of abundance of one or more tRNAs, and special regulation mechanisms, such as attenuation (see Vellanoweth, R. I. (1993) Translation and its regulation in Bacillus subtilis and other gram-positive bacteria, Sonenshein, A. L. et al., eds. ASM: Washington, D.C., p. 699-711 and references cited therein).
  • Transcriptional and translational regulation may be targeted to a single protein (sequential regulation) or simultaneously to several proteins in different metabolic pathways (coordinate regulation). Often, genes whose expression is coordinately regulated are physically located near one another in the genome, in an operon or regulon. Such up- or down-regulation of gene transcription and translation is governed by the cellular and extracellular levels of various factors, such as substrates (precursor and intermediate molecules used in one or more metabolic pathways), catabolites (molecules produced by biochemical pathways concerned with the production of energy from the breakdown of complex organic molecules such as sugars), and end products (the molecules resulting at the end of a metabolic pathway). Typically, the expression of genes encoding enzymes necessary for the activity of a particular pathway is induced by high levels of substrate molecules for that pathway. Similarly, such gene expression tends to be repressed when there exist high intracellular levels of the end product of the pathway (Snyder, L. and Champness, W. (1997) The Molecular Biology of Bacteria ASM: Washington). Gene expression may also be regulated by other external and internal factors, such as environmental conditions (e.g., heat, oxidative stress, or starvation). These global environmental changes cause alterations in the expression of specialized modulating genes, which directly or indirectly (via additional genes or proteins) trigger the expression of genes by means of binding to DNA and thereby inducing or repressing transcription (see, for example, Lin, E. C. C. and Lynch, A. S., eds. (1995) Regulation of Gene Expression in Escherichia coli. Chapman & Hall: New York).
  • Yet another mechanism by which cellular metabolism may be regulated is at the level of the protein. Such regulation is accomplished either by the activities of other proteins, or by binding of low-molecular-weight components which either impede or enable the normal functioning of the protein. Examples of protein regulation by the binding of low-molecular-weight compounds include the binding of GTP or NAD. The binding of a low-molecular-weight chemical is typically reversible, as is the case with the GTP-binding proteins. These proteins exist in two stages (with bound GTP or GDP), one stage being the activated form of the protein, and one stage being inactive.
  • Regulation of protein activity by the action of other enzymes typically takes the form of covalent modification of the protein (i.e., phosphorylation of amino acid residues such as histidine or aspartate, or methylation). Such covalent modification is typically reversible, as mediated by an enzyme of the opposite activity. An example of this is the opposite activities of kinases and phosphorylases in protein phosphorylation; protein kinases phosphorylate specific residues on a target protein (e.g., serine or threonine), while protein phosphorylases remove phosphate groups from such proteins. Typically, enzymes which modulate the activity of other proteins are themselves modulated by external stimuli. These stimuli are mediated through proteins which function as sensors. A well known mechanism by which such sensor proteins may mediate these external signals is by dimerization, but others are also known (see, for example, Msadek, T. et al. (1993) “Two-Component Regulatory Systems”, in: Bacillus subtilis and Other Gram-Positive Bacteria, Sonenshein, A. L. et al., eds., ASM: Washington p. 729-745 and references cited therein).
  • A thorough understanding of the regulatory networks governing cellular metabolism in microorganisms is critical for the high-yield production of chemicals by fermentation. Control systems for the down-regulation of metabolic pathways could be removed or lessened to improve the synthesis of desired chemicals, and similarly, those for the up-regulation of metabolic pathways for a desired product could be constitutively activated or optimized in activity (As shown in Hirose, Y. and Okada, H. (1979) “Microbial Production of Amino Acids”, in: Peppler, H. J. and Perlman, D. (eds.) Microbial Technology 2nd ed. Vol. 1, ch. 7 Academic Press: New York.)
  • III. Elements and Methods of the Invention
  • The present invention is based, at least in part, on the discovery of novel molecules, referred to herein as MR nucleic acid and protein molecules, which regulate, by transcriptional, translational, or post-translational means, one or more metabolic pathways in C. glutamicum. In one embodiment, the MR molecules transcriptionally, translationally, or posttranslationally regulate a metabolic pathway in C. glutamicum. In a preferred embodiment, the activity of the MR molecules of the present invention to regulate one or more C. glutamicum metabolic pathways has an impact on the production of a desired fine chemical by this organism. In a particularly preferred embodiment, the MR molecules of the invention are modulated in activity, such that the C. glutamicum metabolic pathways which the MR proteins of the invention regulate are modulated in efficiency or output, which either directly or indirectly modulates the yield, production, and/or efficiency of production of a desired fine chemical by C. glutamicum.
  • The language, “MR protein” or “MR polypeptide” includes proteins which transcriptionally, translationally, or posttranslationally regulate a metabolic pathway in C. glutamicum. Examples of MR proteins include those encoded by the MR genes set forth in Table 1 and Appendix A. The terms “MR gene” or “MR nucleic acid sequence” include nucleic acid sequences encoding an MR protein, which consist of a coding region and also corresponding untranslated 5′ and 3′ sequence regions. Examples of MR genes include those set forth in Table 1. The terms “production” or “productivity” are art-recognized and include the concentration of the fermentation product (for example, the desired fine chemical) formed within a given time and a given fermentation volume (e.g., kg product per hour per liter). The term “efficiency of production” includes the time required for a particular level of production to be achieved (for example, how long it takes for the cell to attain a particular rate of output of a fine chemical). The term “yield” or “product/carbon yield” is art-recognized and includes the efficiency of the conversion of the carbon source into the product (i.e., fine chemical). This is generally written as, for example, kg product per kg carbon source. By increasing the yield or production of the compound, the quantity of recovered molecules, or of useful recovered molecules of that compound in a given amount of culture over a given amount of time is increased. The terms “biosynthesis” or a “biosynthetic pathway” are art-recognized and include the synthesis of a compound, preferably an organic compound, by a cell from intermediate compounds in what may be a multistep and highly regulated process. The terms “degradation” or a “degradation pathway” are art-recognized and include the breakdown of a compound, preferably an organic compound, by a cell to degradation products (generally speaking, smaller or less complex molecules) in what may be a multistep and highly regulated process. The language “metabolism” is art-recognized and includes the totality of the biochemical reactions that take place in an organism. The metabolism of a particular compound, then, (e.g., the metabolism of an amino acid such as glycine) comprises the overall biosynthetic, modification, and degradation pathways in the cell related to this compound. The term, “regulation” is art-recognized and includes the activity of a protein to govern the activity of another protein. The term, “transcriptional regulation” is art-recognized and includes the activity of a protein to impede or activate the conversion of a DNA encoding a target protein to mRNA. The term, “translational regulation” is art-recognized and includes the activity of a protein to impede or activate the conversion of an mRNA encoding a target protein to a protein molecule. The term, “posttranslational regulation” is art-recognized and includes the activity of a protein to impede or improve the activity of a target protein by covalently modifying the target protein (e.g., by methylation, glucosylation, or phosphorylation).
  • In another embodiment, the MR molecules of the invention are capable of modulating the production of a desired molecule, such as a fine chemical, in a microorganism such as C. glutamicum. Using recombinant genetic techniques, one or more of the regulatory proteins of the invention for metabolic pathways may be manipulated such that its function is modulated. For example, a biosynthetic enzyme may be improved in efficiency, or its allosteric control region destroyed such that feedback inhibition of production of the compound is prevented. Similarly, a degradative enzyme may be deleted or modified by substitution, deletion, or addition such that its degradative activity is lessened for the desired compound without impairing the viability of the cell. In each case, the overall yield or rate of production of one of these desired fine chemicals may be increased.
  • It is also possible that such alterations in the protein and nucleotide molecules of the invention may improve the production of fine chemicals in an indirect fashion. The regulatory mechanisms of metabolic pathways in the cell are necessarily intertwined, and the activation of one pathway may lead to the repression or activation of another in a concomitant fashion. Therefore, by modulating the activity of one or more of the proteins of the invention, the production or efficiency of activity of another fine chemical biosynthetic or degradative pathway may be impacted. For example, by decreasing the ability of an MR protein to repress the transcription of a gene encoding a particular amino acid biosynthetic protein, one may concomitantly derepress other amino acid biosynthetic pathways, since these pathways are interrelated. Further, by modifying the MR proteins of the invention, one may uncouple the growth and division of cells from their extracellular surroundings to a certain degree; by impairing an MR protein which normally represses biosynthesis of a nucleotide when the extracellular conditions are suboptimal for growth and cell division such that it now lacks this function, one may permit growth to occur even when the extracellular conditions are poor. This is of particular relevance in large-scale fermentative growth, where conditions within the culture are often suboptimal in terms of temperature, nutrient supply or aeration, but would still support growth and cell division if the cellular regulatory systems for these factors were eliminated.
  • The isolated nucleic acid sequences of the invention are contained within the genome of a Corynebacterium glutamicum strain available through the American Type Culture Collection, given designation ATCC 13032. The nucleotide sequence of the isolated C. glutamicum MR DNAs and the predicted amino acid sequences of the C. glutamicum MR proteins are shown in Appendices A and B, respectively. Computational analyses were performed which classified and/or identified these nucleotide sequences as sequences which encode metabolic pathway regulatory proteins.
  • The present invention also pertains to proteins which have an amino acid sequence which is substantially homologous to an amino acid sequence of Appendix B. As used herein, a protein which has an amino acid sequence which is substantially homologous to a selected amino acid sequence is least about 50% homologous to the selected amino acid sequence, e.g., the entire selected amino acid sequence. A protein which has an amino acid sequence which is substantially homologous to a selected amino acid sequence can also be least about 50-60%, preferably at least about 60-70%. and more preferably at least about 70-80%, 80-90%, or 90-95%, and most preferably at least about 96%, 97%, 98%, 99% or more homologous to the selected amino acid sequence.
  • The MR protein or a biologically active portion or fragment thereof of the invention can transcriptionally, translationally, or posttranslationally regulate a metabolic pathway in C. glutamicum, or have one or more of the activities set forth in Table 1.
  • Various aspects of the invention are described in further detail in the following subsections:
  • A. Isolated Nucleic Acid Molecules
  • One aspect of the invention pertains to isolated nucleic acid molecules that encode MR polypeptides or biologically active portions thereof, as well as nucleic acid fragments sufficient for use as hybridization probes or primers for the identification or amplification of MR-encoding nucleic acid (e.g., MR DNA). As used herein, the term “nucleic acid molecule” is intended to include DNA molecules (e.g., cDNA or genomic DNA) and RNA molecules (e.g., mRNA) and analogs of the DNA or RNA generated using nucleotide analogs. This term also encompasses untranslated sequence located at both the 3′ and 5′ ends of the coding region of the gene: at least about 100 nucleotides of sequence upstream from the 5′ end of the coding region and at least about 20 nucleotides of sequence downstream from the 3′ end of the coding region of the gene. The nucleic acid molecule can be single-stranded or double-stranded, but preferably is double-stranded DNA. An “isolated” nucleic acid molecule is one which is separated from other nucleic acid molecules which are present in the natural source of the nucleic acid. Preferably, an “isolated” nucleic acid is free of sequences which naturally flank the nucleic acid (i.e., sequences located at the 5′ and 3′ ends of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived. For example, in various embodiments, the isolated MR nucleic acid molecule can contain less than about 5 kb, 4 kb, 3 kb, 2 kb, 1 kb, 0.5 kb or 0.1 kb of nucleotide sequences which naturally flank the nucleic acid molecule in genomic DNA of the cell from which the nucleic acid is derived (e.g., a C. glutamicum cell). Moreover, an “isolated” nucleic acid molecule, such as a DNA molecule, can be substantially free of other cellular material, or culture medium when produced by recombinant techniques, or chemical precursors or other chemicals when chemically synthesized.
  • A nucleic acid molecule of the present invention, e.g., a nucleic acid molecule having a nucleotide sequence of Appendix A, or a portion thereof, can be isolated using standard molecular biology techniques and the sequence information provided herein. For example, a C. glutamicum MR DNA can be isolated from a C. glutamicum library using all or portion of one of the sequences of Appendix A as a hybridization probe and standard hybridization techniques (e.g., as described in Sambrook, J., Fritsh, E. F., and Maniatis, T. Molecular Cloning: A Laboratory Manual. 2nd, ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989). Moreover, a nucleic acid molecule encompassing all or a portion of one of the sequences of Appendix A can be isolated by the polymerase chain reaction using oligonucleotide primers designed based upon this sequence (e.g., a nucleic acid molecule encompassing all or a portion of one of the sequences of Appendix A can be isolated by the polymerase chain reaction using oligonucleotide primers designed based upon this same sequence of Appendix A). For example, mRNA can be isolated from normal endothelial cells (e.g., by the guanidinium-thiocyanate extraction procedure of Chirgwin et al. (1979) Biochemistry 18: 5294-5299) and DNA can be prepared using reverse transcriptase (e.g., Moloney MLV reverse transcriptase, available from Gibco/BRL, Bethesda, Md.; or AMV reverse transcriptase, available from Seikagaku America, Inc., St. Petersburg, Fla.). Synthetic oligonucleotide primers for polymerase chain reaction amplification can be designed based upon one of the nucleotide sequences shown in Appendix A. A nucleic acid of the invention can be amplified using cDNA or, alternatively, genomic DNA, as a template and appropriate oligonucleotide primers according to standard PCR amplification techniques. The nucleic acid so amplified can be cloned into an appropriate vector and characterized by DNA sequence analysis. Furthermore, oligonucleotides corresponding to an MR nucleotide sequence can be prepared by standard synthetic techniques, e.g., using an automated DNA synthesizer.
  • In a preferred embodiment, an isolated nucleic acid molecule of the invention comprises one of the nucleotide sequences shown in Appendix A. The sequences of Appendix A correspond to the Corynebacterium glutamicum MR DNAs of the invention. This DNA comprises sequences encoding MR proteins (i.e., the “coding region”, indicated in each sequence in Appendix A), as well as 5′ untranslated sequences and 3′ untranslated sequences, also indicated in Appendix A. Alternatively, the nucleic acid molecule can comprise only the coding region of any of the sequences in Appendix A.
  • For the purposes of this application, it will be understood that each of the sequences set forth in Appendix A has an identifying RXA, RXN, or RXS number having the designation “RXA”, “RXN”, or “RXS” followed by 5 digits (i.e., RXA00603, RXN03181, or RXS00686). Each of these sequences comprises up to three parts: a 5′ upstream region, a coding region, and a downstream region. Each of these three regions is identified by the same RXA, RXN, or RXS designation to eliminate confusion. The recitation “one of the sequences in Appendix A”, then, refers to any of the sequences in Appendix A, which may be distinguished by their differing RXA, RXN, or RXS designations. The coding region of each of these sequences is translated into a corresponding amino acid sequence, which is set forth in Appendix B. The sequences of Appendix B are identified by the same RXA, RXN, or RXS designations as Appendix A, such that they can be readily correlated. For example, the amino acid sequences in Appendix B designated RXA00603, RXN03181, and RXS00686 are translations of the coding regions of the nucleotide sequence of nucleic acid molecules RXA00603, RXN03181, and RXS00686, respectively, in Appendix A. Each of the RXA, RXN, and RXS nucleotide and amino acid sequences of the invention has also been assigned a SEQ ID NO, as indicated in Table 1. For example, as shown in Table 1, the nucleotide sequence of RXA00603 is SEQ ID NO:5 and the amino acid sequence of RXA00603 is SEQ ID NO: 6.
  • Several of the genes of the invention are “F-designated genes”. An F-designated gene includes those genes set forth in Table 1 which have an ‘F’ in front of the RXA, RXN, or RXS designation. For example, SEQ ID NO:3, designated, as indicated on Table 1, as “F RXA02880”, is an F-designated gene, as are SEQ ID NOs: 21, 27, and 33 (designated on Table 1 as “F RXA02493”, “F RXA00291”, and “F RXA00651”, respectively).
  • In one embodiment, the nucleic acid molecules of the present invention are not intended to include those compiled in Table 2. In the case of the dapD gene, a sequence for this gene was published in Wehrmann, A., et al. (1998) J. Bacteriol. 180(12): 3159-3165. However, the sequence obtained by the inventors of the present application is significantly longer than the published version. It is believed that the published version relied on an incorrect start codon, and thus represents only a fragment of the actual coding region.
  • In another preferred embodiment, an isolated nucleic acid molecule of the invention comprises a nucleic acid molecule which is a complement of one of the nucleotide sequences shown in Appendix A, or a portion thereof. A nucleic acid molecule which is complementary to one of the nucleotide sequences shown in Appendix A is one which is sufficiently complementary to one of the nucleotide sequences shown in Appendix A such that it can hybridize to one of the nucleotide sequences shown in Appendix A, thereby forming a stable duplex.
  • In still another preferred embodiment, an isolated nucleic acid molecule of the invention comprises a nucleotide sequence which is at least about 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, or 60%, preferably at least about 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, or 70%, more preferably at least about 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, or 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, or 90%, or 91%, 92%, 93%, 94%, and even more preferably at least about 95%, 96%, 97%, 98%, 99% or more homologous to a nucleotide sequence shown in Appendix A, or a portion thereof. Ranges and identity values intermediate to the above-recited ranges, (e.g., 70-90% identical or 80-95% identical) are also intended to be encompassed by the present invention. For example, ranges of identity values using a combination of any of the above values recited as upper and/or lower limits are intended to be included. In an additional preferred embodiment, an isolated nucleic acid molecule of the invention comprises a nucleotide sequence which hybridizes, e.g., hybridizes under stringent conditions, to one of the nucleotide sequences shown in Appendix A, or a portion thereof.
  • Moreover, the nucleic acid molecule of the invention can comprise only a portion of the coding region of one of the sequences in Appendix A, for example a fragment which can be used as a probe or primer or a fragment encoding a biologically active portion of an MR protein. The nucleotide sequences determined from the cloning of the MR genes from C. glutamicum allows for the generation of probes and primers designed for use in identifying and/or cloning MR homologues in other cell types and organisms, as well as MR homologues from other Corynebacteria or related species. The probe/primer typically comprises substantially purified oligonucleotide. The oligonucleotide typically comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least about 12, preferably about 25, more preferably about 40, 50 or 75 consecutive nucleotides of a sense strand of one of the sequences set forth in Appendix A, an anti-sense sequence of one of the sequences set forth in Appendix A, or naturally occurring mutants thereof. Primers based on a nucleotide sequence of Appendix A can be used in PCR reactions to clone MR homologues. Probes based on the MR nucleotide sequences can be used to detect transcripts or genomic sequences encoding the same or homologous proteins. In preferred embodiments, the probe further comprises a label group attached thereto, e.g. the label group can be a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor. Such probes can be used as a part of a diagnostic test kit for identifying cells which misexpress an MR protein, such as by measuring a level of an MR-encoding nucleic acid in a sample of cells, e.g., detecting MR mRNA levels or determining whether a genomic MR gene has been mutated or deleted.
  • In one embodiment, the nucleic acid molecule of the invention encodes a protein or portion thereof which includes an amino acid sequence which is sufficiently homologous to an amino acid sequence of Appendix B such that the protein or portion thereof maintains the ability to transcriptionally, translationally, or posttranslationally regulate a metabolic pathway in C. glutamicum. As used herein, the language “sufficiently homologous” refers to proteins or portions thereof which have amino acid sequences which include a minimum number of identical or equivalent (e.g., an amino acid residue which has a similar side chain as an amino acid residue in one of the sequences of Appendix B) amino acid residues to an amino acid sequence of Appendix B such that the protein or portion thereof is able to transcriptionally, translationally, or posttranslationally regulate a metabolic pathway in C. glutamicum. Protein members of such metabolic pathways, as described herein, may function to regulate the biosynthesis or degradation of one or more fine chemicals. Examples of such activities are also described herein. Thus, “the function of an MR protein” contributes to the overall regulation of one or more fine chemical metabolic pathway, or contributes, either directly or indirectly, to the yield, production, and/or efficiency of production of one or more fine chemicals. Examples of MR protein activities are set forth in Table 1.
  • In another embodiment, the protein is at least about 50-60%, preferably at least about 60-70%, and more preferably at least about 70-80%, 80-90%, 90-95%, and most preferably at least about 96%, 97%, 98%, 99% or more homologous to an entire amino acid sequence of Appendix B.
  • Portions of proteins encoded by the MR nucleic acid molecules of the invention are preferably biologically active portions of one of the MR proteins. As used herein, the term “biologically active portion of an MR protein” is intended to include a portion, e.g., a domain/motif, of an MR protein that transcriptionally, translationally, or posttranslationally regulates a metabolic pathway in C. glutamicum, or has an activity as set forth in Table 1. To determine whether an MR protein or a biologically active portion thereof can transcriptionally, translationally, or posttranslationally regulate a metabolic pathway in C. glutamicum, an assay of enzymatic activity may be performed. Such assay methods are well known to those of ordinary skill in the art, as detailed in Example 8 of the Exemplification.
  • Additional nucleic acid fragments encoding biologically active portions of an MR protein can be prepared by isolating a portion of one of the sequences in Appendix B, expressing the encoded portion of the MR protein or peptide (e.g., by recombinant expression in vitro) and assessing the activity of the encoded portion of the MR protein or peptide.
  • The invention further encompasses nucleic acid molecules that differ from one of the nucleotide sequences shown in Appendix A (and portions thereof) due to degeneracy of the genetic code and thus encode the same MR protein as that encoded by the nucleotide sequences shown in Appendix A. In another embodiment, an isolated nucleic acid molecule of the invention has a nucleotide sequence encoding a protein having an amino acid sequence shown in Appendix B. In a still further embodiment, the nucleic acid molecule of the invention encodes a full length C. glutamicum protein which is substantially homologous to an amino acid sequence of Appendix B (encoded by an open reading frame shown in Appendix A).
  • It will be understood by one of ordinary skill in the art that in one embodiment the sequences of the invention are not meant to include the sequences of the prior art, such as those Genbank sequences set forth in Tables 2 or 4 which were available prior to the present invention. In one embodiment, the invention includes nucleotide and amino acid sequences having a percent identity to a nucleotide or amino acid sequence of the invention which is greater than that of a sequence of the prior art (e.g., a Genbank sequence (or the protein encoded by such a sequence) set forth in Tables 2 or 4). For example, the invention includes a nucleotide sequence which is greater than and/or at least 40% identical to the nucleotide sequence designated RXA00603 (SEQ ID NO:5), a nucleotide sequence which is greater than and/or at least 55% identical to the nucleotide sequence designated RXA00129 (SEQ ID NO:29), and a nucleotide sequence which is greater than and/or at least 40% identical to the nucleotide sequence designated RXA00006 (SEQ ID NO:35). One of ordinary skill in the art would be able to calculate the lower threshold of percent identity for any given sequence of the invention by examining the GAP-calculated percent identity scores set forth in Table 4 for each of the three top hits for the given sequence, and by subtracting the highest GAP-calculated percent identity from 100 percent. One of ordinary skill in the art will also appreciate that nucleic acid and amino acid sequences having percent identities greater than the lower threshold so calculated (e.g., at least 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, or 60%, preferably at least about 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, or 70%, more preferably at least about 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, or 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, or 90%, or 91%, 92%, 93%, 94%, and even more preferably at least about 95%, 96%, 97%, 98%, 99% or more identical) are also encompassed by the invention.
  • In addition to the C. glutamicum MR nucleotide sequences shown in Appendix A, it will be appreciated by those of ordinary skill in the art that DNA sequence polymorphisms that lead to changes in the amino acid sequences of MR proteins may exist within a population (e.g., the C. glutamicum population). Such genetic polymorphism in the MR gene may exist among individuals within a population due to natural variation. As used herein, the terms “gene” and “recombinant gene” refer to nucleic acid molecules comprising an open reading frame encoding an MR protein, preferably a C. glutamicum MR protein. Such natural variations can typically result in 1-5% variance in the nucleotide sequence of the MR gene. Any and all such nucleotide variations and resulting amino acid polymorphisms in MR that are the result of natural variation and that do not alter the functional activity of MR proteins are intended to be within the scope of the invention.
  • Nucleic acid molecules corresponding to natural variants and non-C. glutamicum homologues of the C. glutamicum MR DNA of the invention can be isolated based on their homology to the C. glutamicum MR nucleic acid disclosed herein using the C. glutamicum DNA, or a portion thereof, as a hybridization probe according to standard hybridization techniques under stringent hybridization conditions. Accordingly, in another embodiment, an isolated nucleic acid molecule of the invention is at least 15 nucleotides in length and hybridizes under stringent conditions to the nucleic acid molecule comprising a nucleotide sequence of Appendix A. In other embodiments, the nucleic acid is at least 30, 50, 100, 250 or more nucleotides in length. As used herein, the term “hybridizes under stringent conditions” is intended to describe conditions for hybridization and washing under which nucleotide sequences at least 60% homologous to each other typically remain hybridized to each other. Preferably, the conditions are such that sequences at least about 65%, more preferably at least about 70%, and even more preferably at least about 75% or more homologous to each other typically remain hybridized to each other. Such stringent conditions are known to those of ordinary skill in the art and can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6. A preferred, non-limiting example of stringent hybridization conditions are hybridization in 6× sodium chloride/sodium citrate (SSC) at about 45° C., followed by one or more washes in 0.2×SSC, 0.1% SDS at 50-65° C. Preferably, an isolated nucleic acid molecule of the invention that hybridizes under stringent conditions to a sequence of Appendix A corresponds to a naturally-occurring nucleic acid molecule. As used herein, a “naturally-occurring” nucleic acid molecule refers to an RNA or DNA molecule having a nucleotide sequence that occurs in nature (e.g., encodes a natural protein). In one embodiment, the nucleic acid encodes a natural C. glutamicum MR protein.
  • In addition to naturally-occurring variants of the MR sequence that may exist in the population, one of ordinary skill in the art will further appreciate that changes can be introduced by mutation into a nucleotide sequence of Appendix A, thereby leading to changes in the amino acid sequence of the encoded MR protein, without altering the functional ability of the MR protein. For example, nucleotide substitutions leading to amino acid substitutions at “non-essential” amino acid residues can be made in a sequence of Appendix A. A “non-essential” amino acid residue is a residue that can be altered from the wild-type sequence of one of the MR proteins (Appendix B) without altering the activity of said MR protein, whereas an “essential” amino acid residue is required for MR protein activity. Other amino acid residues, however, (e.g., those that are not conserved or only semi-conserved in the domain having MR activity) may not be essential for activity and thus are likely to be amenable to alteration without altering MR activity.
  • Accordingly, another aspect of the invention pertains to nucleic acid molecules encoding MR proteins that contain changes in amino acid residues that are not essential for MR activity. Such MR proteins differ in amino acid sequence from a sequence contained in Appendix B yet retain at least one of the MR activities described herein. In one embodiment, the isolated nucleic acid molecule comprises a nucleotide sequence encoding a protein, wherein the protein comprises an amino acid sequence at least about 50% homologous to an amino acid sequence of Appendix B and is capable of transcriptionally, translationally, or posttranslationally regulating a metabolic pathway in C. glutamicum, or has one or more activities set forth in Table 1. Preferably, the protein encoded by the nucleic acid molecule is at least about 50-60% homologous to one of the sequences in Appendix B, more preferably at least about 60-70% homologous to one of the sequences in Appendix B, even more preferably at least about 70-80%, 80-90%, 90-95% homologous to one of the sequences in Appendix B, and most preferably at least about 96%, 97%, 98%, or 99% homologous to one of the sequences in Appendix B.
  • To determine the percent homology of two amino acid sequences (e.g., one of the sequences of Appendix B and a mutant form thereof) or of two nucleic acids, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of one protein or nucleic acid for optimal alignment with the other protein or nucleic acid). The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in one sequence (e.g., one of the sequences of Appendix B) is occupied by the same amino acid residue or nucleotide as the corresponding position in the other sequence (e.g., a mutant form of the sequence selected from Appendix B), then the molecules are homologous at that position (i.e., as used herein amino acid or nucleic acid “homology” is equivalent to amino acid or nucleic acid “identity”). The percent homology between the two sequences is a function of the number of identical positions shared by the sequences (i.e., % homology=# of identical positions/total # of positions×100).
  • An isolated nucleic acid molecule encoding an MR protein homologous to a protein sequence of Appendix B can be created by introducing one or more nucleotide substitutions, additions or deletions into a nucleotide sequence of Appendix A such that one or more amino acid substitutions, additions or deletions are introduced into the encoded protein. Mutations can be introduced into one of the sequences of Appendix A by standard techniques, such as site-directed mutagenesis and PCR-mediated mutagenesis. Preferably, conservative amino acid substitutions are made at one or more predicted non-essential amino acid residues. A “conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Thus, a predicted nonessential amino acid residue in an MR protein is preferably replaced with another amino acid residue from the same side chain family. Alternatively, in another embodiment, mutations can be introduced randomly along all or part of an MR coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for an MR activity described herein to identify mutants that retain MR activity. Following mutagenesis of one of the sequences of Appendix A, the encoded protein can be expressed recombinantly and the activity of the protein can be determined using, for example, assays described herein (see Example 8 of the Exemplification).
  • In addition to the nucleic acid molecules encoding MR proteins described above, another aspect of the invention pertains to isolated nucleic acid molecules which are antisense thereto. An “antisense” nucleic acid comprises a nucleotide sequence which is complementary to a “sense” nucleic acid encoding a protein, e.g., complementary to the coding strand of a double-stranded DNA molecule or complementary to an mRNA sequence. Accordingly, an antisense nucleic acid can hydrogen bond to a sense nucleic acid. The antisense nucleic acid can be complementary to an entire MR coding strand, or to only a portion thereof. In one embodiment, an antisense nucleic acid molecule is antisense to a “coding region” of the coding strand of a nucleotide sequence encoding an MR protein. The term “coding region” refers to the region of the nucleotide sequence comprising codons which are translated into amino acid residues (e.g., the entire coding region of SEQ ID NO: 1 (RXN03181) comprises nucleotides 1 to 414). In another embodiment, the antisense nucleic acid molecule is antisense to a “noncoding region” of the coding strand of a nucleotide sequence encoding MR. The term “noncoding region” refers to 5′ and 3′ sequences which flank the coding region that are not translated into amino acids (i.e., also referred to as 5′ and 3′ untranslated regions).
  • Given the coding strand sequences encoding MR disclosed herein (e.g., the sequences set forth in Appendix A), antisense nucleic acids of the invention can be designed according to the rules of Watson and Crick base pairing. The antisense nucleic acid molecule can be complementary to the entire coding region of MR mRNA, but more preferably is an oligonucleotide which is antisense to only a portion of the coding or noncoding region of MR mRNA. For example, the antisense oligonucleotide can be complementary to the region surrounding the translation start site of MR mRNA. An antisense oligonucleotide can be, for example, about 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 nucleotides in length. An antisense nucleic acid of the invention can be constructed using chemical synthesis and enzymatic ligation reactions using procedures known in the art. For example, an antisense nucleic acid (e.g., an antisense oligonucleotide) can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids, e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used. Examples of modified nucleotides which can be used to generate the antisense nucleic acid include 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl)uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D-mannosylqueosine, 5′-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N-6-isopentenyladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5-oxyacetic acid methylester, uracil-5-oxyacetic acid (v), 5-methyl-2-thiouracil, 3-(3-amino-3-N-2-carboxypropyl)uracil, (acp3)w, and 2,6-diaminopurine. Alternatively, the antisense nucleic acid can be produced biologically using an expression vector into which a nucleic acid has been subcloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest, described further in the following subsection).
  • The antisense nucleic acid molecules of the invention are typically administered to a cell or generated in situ such that they hybridize with or bind to cellular mRNA and/or genomic DNA encoding an MR protein to thereby inhibit expression of the protein, e.g., by inhibiting transcription and/or translation. The hybridization can be by conventional nucleotide complementarity to form a stable duplex, or, for example, in the case of an antisense nucleic acid molecule which binds to DNA duplexes, through specific interactions in the major groove of the double helix. The antisense molecule can be modified such that it specifically binds to a receptor or an antigen expressed on a selected cell surface, e.g., by linking the antisense nucleic acid molecule to a peptide or an antibody which binds to a cell surface receptor or antigen. The antisense nucleic acid molecule can also be delivered to cells using the vectors described herein. To achieve sufficient intracellular concentrations of the antisense molecules, vector constructs in which the antisense nucleic acid molecule is placed under the control of a strong prokaryotic, viral, or eukaryotic promoter are preferred.
  • In yet another embodiment, the antisense nucleic acid molecule of the invention is an α-anomeric nucleic acid molecule. An α-anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual β-units, the strands run parallel to each other (Gaultier et al. (1987) Nucleic Acids. Res. 15:6625-6641). The antisense nucleic acid molecule can also comprise a 2′-o-methylribonucleotide (Inoue et al. (1987) Nucleic Acids Res. 15:6131-6148) or a chimeric RNA-DNA analogue (Inoue et al. (1987) FEBS Lett. 215:327-330).
  • In still another embodiment, an antisense nucleic acid of the invention is a ribozyme. Ribozymes are catalytic RNA molecules with ribonuclease activity which are capable of cleaving a single-stranded nucleic acid, such as an mRNA, to which they have a complementary region. Thus, ribozymes (e.g., hammerhead ribozymes (described in Haselhoff and Gerlach (1988) Nature 334:585-591)) can be used to catalytically cleave MR mRNA transcripts to thereby inhibit translation of MR mRNA. A ribozyme having specificity for an MR-encoding nucleic acid can be designed based upon the nucleotide sequence of an MR DNA disclosed herein (i.e., SEQ ID NO:1 (RXN03181 in Appendix A)). For example, a derivative of a Tetrahymena L-19 IVS RNA can be constructed in which the nucleotide sequence of the active site is complementary to the nucleotide sequence to be cleaved in an MR-encoding mRNA. See, e.g., Cech et al. U.S. Pat. No. 4,987,071 and Cech et al. U.S. Pat. No. 5,116,742. Alternatively, MR mRNA can be used to select a catalytic RNA having a specific ribonuclease activity from a pool of RNA molecules. See, e.g., Bartel, D. and Szostak, J. W. (1993) Science 261:1411-1418.
  • Alternatively, MR gene expression can be inhibited by targeting nucleotide sequences complementary to the regulatory region of an MR nucleotide sequence (e.g., an MR promoter and/or enhancers) to form triple helical structures that prevent transcription of an MR gene in target cells. See generally, Helene, C. (1991) Anticancer Drug Des. 6(6):569-84; Helene, C. et al. (1992) Ann. N.Y. Acad. Sci. 660:27-36; and Maher, L. J. (1992) Bioassays 14(12):807-15.
  • B. Recombinant Expression Vectors and Host Cells
  • Another aspect of the invention pertains to vectors, preferably expression vectors, containing a nucleic acid encoding an MR protein (or a portion thereof). As used herein, the term “vector” refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a “plasmid”, which refers to a circular double stranded DNA loop into which additional DNA segments can be ligated. Another type of vector is a viral vector, wherein additional DNA segments can be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as “expression vectors”. In general, expression vectors of utility in recombinant DNA techniques are often in the form of plasmids. In the present specification, “plasmid” and “vector” can be used interchangeably as the plasmid is the most commonly used form of vector. However, the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions.
  • The recombinant expression vectors of the invention comprise a nucleic acid of the invention in a form suitable for expression of the nucleic acid in a host cell, which means that the recombinant expression vectors include one or more regulatory sequences, selected on the basis of the host cells to be used for expression, which is operatively linked to the nucleic acid sequence to be expressed. Within a recombinant expression vector, “operably linked” is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner which allows for expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell). The term “regulatory sequence” is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel; Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990). Regulatory sequences include those which direct constitutive expression of a nucleotide sequence in many types of host cell and those which direct expression of the nucleotide sequence only in certain host cells. Preferred regulatory sequences are, for example, promoters such as cos-, tac-, trp-, tet-, trp-tet-, lpp-, lac-, lpp-lac-, lacIq-, T7-, T5-, T3-, gal-, trc-, ara-, SP6-, arny, SPO2, λ-PR- or λPL, which are used preferably in bacteria. Additional regulatory sequences are, for example, promoters from yeasts and fungi, such as ADC1, MFα, AC, P-60, CYC1, GAPDH, TEF, rp28, ADH, promoters from plants such as CaMV/35S, SSU, OCS, lib4, usp, STLS1, B33, nos or ubiquitin- or phaseolin-promoters. It is also possible to use artificial promoters. It will be appreciated by one of ordinary skill in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc. The expression vectors of the invention can be introduced into host cells to thereby produce proteins or peptides, including fusion proteins or peptides, encoded by nucleic acids as described herein (e.g., MR proteins, mutant forms of MR proteins, fusion proteins, etc.).
  • The recombinant expression vectors of the invention can be designed for expression of MR proteins in prokaryotic or eukaryotic cells. For example, MR genes can be expressed in bacterial cells such as C. glutamicum, insect cells (using baculovirus expression vectors), yeast and other fungal cells (see Romanos, M. A. et al. (1992) “Foreign gene expression in yeast: a review”, Yeast 8: 423-488; van den Hondel, C. A. M. J. J. et al. (1991) “Heterologous gene expression in filamentous fungi” in: More Gene Manipulations in Fungi, J. W. Bennet & L. L. Lasure, eds., p. 396-428: Academic Press: San Diego; and van den Hondel, C. A. M. J. J. & Punt, P. J. (1991) “Gene transfer systems and vector development for filamentous fungi, in: Applied Molecular Genetics of Fungi, Peberdy, J. F. et al., eds., p. 1-28, Cambridge University Press: Cambridge), algae and multicellular plant cells (see Schmidt, R. and Willmitzer, L. (1988) High efficiency Agrobacterium tumefaciens-mediated transformation of Arabidopsis thaliana leaf and cotyledon explants” Plant Cell Rep.: 583-586), or mammalian cells. Suitable host cells are discussed further in Goeddel, Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990). Alternatively, the recombinant expression vector can be transcribed and translated in vitro, for example using T7 promoter regulatory sequences and T7 polymerase.
  • Expression of proteins in prokaryotes is most often carried out with vectors containing constitutive or inducible promoters directing the expression of either fusion or non-fusion proteins. Fusion vectors add a number of amino acids to a protein encoded therein, usually to the amino terminus of the recombinant protein. Such fusion vectors typically serve three purposes: 1) to increase expression of recombinant protein; 2) to increase the solubility of the recombinant protein; and 3) to aid in the purification of the recombinant protein by acting as a ligand in affinity purification. Often, in fusion expression vectors, a proteolytic cleavage site is introduced at the junction of the fusion moiety and the recombinant protein to enable separation of the recombinant protein from the fusion moiety subsequent to purification of the fusion protein. Such enzymes, and their cognate recognition sequences, include Factor Xa, thrombin and enterokinase.
  • Typical fusion expression vectors include pGEX (Pharmacia Biotech Inc; Smith, D. B. and Johnson, K. S. (1988) Gene 67:31-40), pMAL (New England Biolabs, Beverly, Mass.) and pRIT5 (Pharmacia, Piscataway, N.J.) which fuse glutathione S-transferase (GST), maltose E binding protein, or protein A, respectively, to the target recombinant protein. In one embodiment, the coding sequence of the MR protein is cloned into a pGEX expression vector to create a vector encoding a fusion protein comprising, from the N-terminus to the C-terminus, GST-thrombin cleavage site-X protein. The fusion protein can be purified by affinity chromatography using glutathione-agarose resin. Recombinant MR protein unfused to GST can be recovered by cleavage of the fusion protein with thrombin.
  • Examples of suitable inducible non-fusion E. coli expression vectors include pTrc (Amann et al., (1988) Gene 69:301-315) pLG338, pACYC184, pBR322, pUC18, pUC19, pKC30, pRep4, pHS1, pHS2, pPLc236, pMBL24, pLG200, pUR290, pIN-III113-B1, λgt11, pBdCl, and pET 11d (Studier et al., Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990) 60-89; and Pouwels et al., eds. (1985) Cloning Vectors. Elsevier: New York IBSN 0 444 904018). Target gene expression from the pTrc vector relies on host RNA polymerase transcription from a hybrid trp-lac fusion promoter. Target gene expression from the pET 11d vector relies on transcription from a T7 gn10-lac fusion promoter mediated by a coexpressed viral RNA polymerase (T7 gn1). This viral polymerase is supplied by host strains BL21(DE3) or HMS174(DE3) from a resident λ prophage harboring a T7 gn1 gene under the transcriptional control of the lacUV 5 promoter. For transformation of other varieties of bacteria, appropriate vectors may be selected. For example, the plasmids pIJ101, pIJ364, pIJ702 and pIJ361 are known to be useful in transforming Streptomyces, while plasmids pUB110, pC194, or pBD214 are suited for transformation of Bacillus species. Several plasmids of use in the transfer of genetic information into Corynebacterium include pHM1519, pBL1, pSA77, or pAJ667 (Pouwels et al., eds. (1985) Cloning Vectors. Elsevier: New York IBSN 0 444 904018). One strategy to maximize recombinant protein expression is to express the protein in a host bacteria with an impaired capacity to proteolytically cleave the recombinant protein (Gottesman, S., Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990) 119-128). Another strategy is to alter the nucleic acid sequence of the nucleic acid to be inserted into an expression vector so that the individual codons for each amino acid are those preferentially utilized in the bacterium chosen for expression, such as C. glutamicum (Wada et al. (1992) Nucleic Acids Res. 20:2111-2118). Such alteration of nucleic acid sequences of the invention can be carried out by standard DNA synthesis techniques.
  • In another embodiment, the MR protein expression vector is a yeast expression vector. Examples of vectors for expression in yeast S. cerevisiae include pYepSec1 (Baldari, et al., (1987) Embo J. 6:229-234), 2μ, pAG-1, Yep6, Yep13, pEMBLYe23, pMFa (Kurjan and Herskowitz, (1982) Cell 30:933-943), pJRY88 (Schultz et al., (1987) Gene 54:113-123), and pYES2 (Invitrogen Corporation, San Diego, Calif.). Vectors and methods for the construction of vectors appropriate for use in other fungi, such as the filamentous fungi, include those detailed in: van den Hondel, C. A. M. J. J. & Punt, P. J. (1991) “Gene transfer systems and vector development for filamentous fungi, in: Applied Molecular Genetics of Fungi, J. F. Peberdy, et al., eds., p. 1-28, Cambridge University Press: Cambridge, and Pouwels et al., eds. (1985) Cloning Vectors. Elsevier: New York (IBSN 0 444 904018).
  • Alternatively, the MR proteins of the invention can be expressed in insect cells using baculovirus expression vectors. Baculovirus vectors available for expression of proteins in cultured insect cells (e.g., Sf 9 cells) include the pAc series (Smith et al. (1983) Mol. Cell Biol. 3:2156-2165) and the pVL series (Lucklow and Summers (1989) Virology 170:31-39).
  • In another embodiment, the MR proteins of the invention may be expressed in unicellular plant cells (such as algae) or in plant cells from higher plants (e.g., the spermatophytes, such as crop plants). Examples of plant expression vectors include those detailed in: Becker, D., Kemper, E., Schell, J. and Masterson, R. (1992) “New plant binary vectors with selectable markers located proximal to the left border”, Plant Mol. Biol. 20: 1195-1197; and Bevan, M. W. (1984) “Binary Agrobacterium vectors for plant transformation”, Nucl. Acid. Res. 12: 8711-8721, and include pLGV23, pGHlac+, pBIN19, pAK2004, and pDH51 (Pouwels et al., eds. (1985) Cloning Vectors. Elsevier: New York IBSN 0 444 904018).
  • In yet another embodiment, a nucleic acid of the invention is expressed in mammalian cells using a mammalian expression vector. Examples of mammalian expression vectors include pCDM8 (Seed, B. (1987) Nature 329:840) and pMT2PC (Kaufman et al. (1987) EMBO J. 6:187-195). When used in mammalian cells, the expression vector's control functions are often provided by viral regulatory elements. For example, commonly used promoters are derived from polyoma, Adenovirus 2, cytomegalovirus and Simian Virus 40. For other suitable expression systems for both prokaryotic and eukaryotic cells see chapters 16 and 17 of Sambrook, J., Fritsh, E. F., and Maniatis, T. Molecular Cloning: A Laboratory Manual. 2nd, ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989.
  • In another embodiment, the recombinant mammalian expression vector is capable of directing expression of the nucleic acid preferentially in a particular cell type (e.g., tissue-specific regulatory elements are used to express the nucleic acid). Tissue-specific regulatory elements are known in the art. Non-limiting examples of suitable tissue-specific promoters include the albumin promoter (liver-specific; Pinkert et al. (1987) Genes Dev. 1:268-277), lymphoid-specific promoters (Calame and Eaton (1988) Adv. Immunol. 43:235-275), in particular promoters of T cell receptors (Winoto and Baltimore (1989) EMBO J. 8:729-733) and immunoglobulins (Banerji et al. (1983) Cell 33:729-740; Queen and Baltimore (1983) Cell 33:741-748), neuron-specific promoters (e.g., the neurofilament promoter; Byrne and Ruddle (1989) PNAS 86:5473-5477), pancreas-specific promoters (Edlund et al. (1985) Science 230:912-916), and mammary gland-specific promoters (e.g., milk whey promoter; U.S. Pat. No. 4,873,316 and European Application Publication No. 264,166). Developmentally-regulated promoters are also encompassed, for example the murine hox promoters (Kessel and Gruss (1990) Science 249:374-379) and the α-fetoprotein promoter (Campes and Tilghman (1989) Genes Dev. 3:537-546).
  • The invention further provides a recombinant expression vector comprising a DNA molecule of the invention cloned into the expression vector in an antisense orientation. That is, the DNA molecule is operatively linked to a regulatory sequence in a manner which allows for expression (by transcription of the DNA molecule) of an RNA molecule which is antisense to MR mRNA. Regulatory sequences operatively linked to a nucleic acid cloned in the antisense orientation can be chosen which direct the continuous expression of the antisense RNA molecule in a variety of cell types, for instance viral promoters and/or enhancers, or regulatory sequences can be chosen which direct constitutive, tissue specific or cell type specific expression of antisense RNA. The antisense expression vector can be in the form of a recombinant plasmid, phagemid or attenuated virus in which antisense nucleic acids are produced under the control of a high efficiency regulatory region, the activity of which can be determined by the cell type into which the vector is introduced. For a discussion of the regulation of gene expression using antisense genes see Weintraub, H. et al., Antisense RNA as a molecular tool for genetic analysis, Reviews—Trends in Genetics, Vol. 1(1) 1986.
  • Another aspect of the invention pertains to host cells into which a recombinant expression vector of the invention has been introduced. The terms “host cell” and “recombinant host cell” are used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
  • A host cell can be any prokaryotic or eukaryotic cell. For example, an MR protein can be expressed in bacterial cells such as C. glutamicum, insect cells, yeast or mammalian cells (such as Chinese hamster ovary cells (CHO) or COS cells). Other suitable host cells are known to one of ordinary skill in the art. Microorganisms related to Corynebacterium glutamicum which may be conveniently used as host cells for the nucleic acid and protein molecules of the invention are set forth in Table 3.
  • Vector DNA can be introduced into prokaryotic or eukaryotic cells via conventional transformation or transfection techniques. As used herein, the terms “transformation” and “transfection” are intended to refer to a variety of art-recognized techniques for introducing foreign nucleic acid (e.g., linear DNA or RNA (e.g., a linearized vector or a gene construct alone without a vector) or nucleic acid in the form of a vector (e.g., a plasmid, phage, phasmid, phagemid, transposon or other DNA) into a host cell, including calcium phosphate or calcium chloride co-precipitation, DEAE-dextran-mediated transfection, lipofection, or electroporation. Suitable methods for transforming or transfecting host cells can be found in Sambrook, et al. (Molecular Cloning: A Laboratory Manual. 2nd, ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989), and other laboratory manuals.
  • For stable transfection of mammalian cells, it is known that, depending upon the expression vector and transfection technique used, only a small fraction of cells may integrate the foreign DNA into their genome. In order to identify and select these integrants, a gene that encodes a selectable marker (e.g., resistance to antibiotics) is generally introduced into the host cells along with the gene of interest. Preferred selectable markers include those which confer resistance to drugs, such as G418, hygromycin and methotrexate. Nucleic acid encoding a selectable marker can be introduced into a host cell on the same vector as that encoding an MR protein or can be introduced on a separate vector. Cells stably transfected with the introduced nucleic acid can be identified by, for example, drug selection (e.g., cells that have incorporated the selectable marker gene will survive, while the other cells die).
  • To create a homologous recombinant microorganism, a vector is prepared which contains at least a portion of an MR gene into which a deletion, addition or substitution has been introduced to thereby alter, e.g., functionally disrupt, the MR gene. Preferably, this MR gene is a Corynebacterium glutamicum MR gene, but it can be a homologue from a related bacterium or even from a mammalian, yeast, or insect source. In a preferred embodiment, the vector is designed such that, upon homologous recombination, the endogenous MR gene is functionally disrupted (i.e., no longer encodes a functional protein; also referred to as a “knock out” vector). Alternatively, the vector can be designed such that, upon homologous recombination, the endogenous MR gene is mutated or otherwise altered but still encodes functional protein (e.g., the upstream regulatory region can be altered to thereby alter the expression of the endogenous MR protein). In the homologous recombination vector, the altered portion of the MR gene is flanked at its 5′ and 3′ ends by additional nucleic acid of the MR gene to allow for homologous recombination to occur between the exogenous MR gene carried by the vector and an endogenous MR gene in a microorganism. The additional flanking MR nucleic acid is of sufficient length for successful homologous recombination with the endogenous gene. Typically, several kilobases of flanking DNA (both at the 5′ and 3′ ends) are included in the vector (see e.g., Thomas, K. R., and Capecchi, M. R. (1987) Cell 51: 503 for a description of homologous recombination vectors). The vector is introduced into a microorganism (e.g., by electroporation) and cells in which the introduced MR gene has homologously recombined with the endogenous MR gene are selected, using art-known techniques.
  • In another embodiment, recombinant microorganisms can be produced which contain selected systems which allow for regulated expression of the introduced gene. For example, inclusion of an MR gene on a vector placing it under control of the lac operon permits expression of the MR gene only in the presence of IPTG. Such regulatory systems are well known in the art.
  • In another embodiment, an endogenous MR gene in a host cell is disrupted (e.g., by homologous recombination or other genetic means known in the art) such that expression of its protein product does not occur. In another embodiment, an endogenous or introduced MR gene in a host cell has been altered by one or more point mutations, deletions, or inversions, but still encodes a functional MR protein. In still another embodiment, one or more of the regulatory regions (e.g., a promoter, repressor, or inducer) of an MR gene in a microorganism has been altered (e.g., by deletion, truncation, inversion, or point mutation) such that the expression of the MR gene is modulated. One of ordinary skill in the art will appreciate that host cells containing more than one of the described MR gene and protein modifications may be readily produced using the methods of the invention, and are meant to be included in the present invention.
  • A host cell of the invention, such as a prokaryotic or eukaryotic host cell in culture, can be used to produce (i.e., express) an MR protein. Accordingly, the invention further provides methods for producing MR proteins using the host cells of the invention. In one embodiment, the method comprises culturing the host cell of invention (into which a recombinant expression vector encoding an MR protein has been introduced, or into which genome has been introduced a gene encoding a wild-type or altered MR protein) in a suitable medium until MR protein is produced. In another embodiment, the method further comprises isolating MR proteins from the medium or the host cell.
  • C. Isolated MR Proteins
  • Another aspect of the invention pertains to isolated MR proteins, and biologically active portions thereof. An “isolated” or “purified” protein or biologically active portion thereof is substantially free of cellular material when produced by recombinant DNA techniques, or chemical precursors or other chemicals when chemically synthesized. The language “substantially free of cellular material” includes preparations of MR protein in which the protein is separated from cellular components of the cells in which it is naturally or recombinantly produced. In one embodiment, the language “substantially free of cellular material” includes preparations of MR protein having less than about 30% (by dry weight) of non-MR protein (also referred to herein as a “contaminating protein”), more preferably less than about 20% of non-MR protein, still more preferably less than about 10% of non-MR protein, and most preferably less than about 5% non-MR protein. When the MR protein or biologically active portion thereof is recombinantly produced, it is also preferably substantially free of culture medium, i.e., culture medium represents less than about 20%, more preferably less than about 10%, and most preferably less than about 5% of the volume of the protein preparation. The language “substantially free of chemical precursors or other chemicals” includes preparations of MR protein in which the protein is separated from chemical precursors or other chemicals which are involved in the synthesis of the protein. In one embodiment, the language “substantially free of chemical precursors or other chemicals” includes preparations of MR protein having less than about 30% (by dry weight) of chemical precursors or non-MR chemicals, more preferably less than about 20% chemical precursors or non-MR chemicals, still more preferably less than about 10% chemical precursors or non-MR chemicals, and most preferably less than about 5% chemical precursors or non-MR chemicals. In preferred embodiments, isolated proteins or biologically active portions thereof lack contaminating proteins from the same organism from which the MR protein is derived. Typically, such proteins are produced by recombinant expression of, for example, a C. glutamicum MR protein in a microorganism such as C. glutamicum.
  • An isolated MR protein or a portion thereof of the invention can transcriptionally, translationally, or posttranslationally regulate a metabolic pathway in C. glutamicum, or has one or more of the activities set forth in Table 1. In preferred embodiments, the protein or portion thereof comprises an amino acid sequence which is sufficiently homologous to an amino acid sequence of Appendix B such that the protein or portion thereof maintains the ability to transcriptionally, translationally, or posttranslationally regulate a metabolic pathway in C. glutamicum. The portion of the protein is preferably a biologically active portion as described herein. In another preferred embodiment, an MR protein of the invention has an amino acid sequence shown in Appendix B. In yet another preferred embodiment, the MR protein has an amino acid sequence which is encoded by a nucleotide sequence which hybridizes, e.g., hybridizes under stringent conditions, to a nucleotide sequence of Appendix A. In still another preferred embodiment, the MR protein has an amino acid sequence which is encoded by a nucleotide sequence that is at least about 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, or 60%, preferably at least about 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, or 70%, more preferably at least about 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, or 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, or 90%, or 91%, 92%, 93%, 94%, and even more preferably at least about 95%, 96%, 97%, 98%, 99% or more homologous to one of the nucleic acid sequences of Appendix A, or a portion thereof. Ranges and identity values intermediate to the above-recited values, (e.g., 70-90% identical or 80-95% identical) are also intended to be encompassed by the present invention. For example, ranges of identity values using a combination of any of the above values recited as upper and/or lower limits are intended to be included. The preferred MR proteins of the present invention also preferably possess at least one of the MR activities described herein. For example, a preferred MR protein of the present invention includes an amino acid sequence encoded by a nucleotide sequence which hybridizes, e.g., hybridizes under stringent conditions, to a nucleotide sequence of Appendix A, and which can transcriptionally, translationally, or posttranslationally regulate a metabolic pathway in C. glutamicum, or which has one or more of the activities set forth in Table 1.
  • In other embodiments, the MR protein is substantially homologous to an amino acid sequence of Appendix B and retains the functional activity of the protein of one of the sequences of Appendix B yet differs in amino acid sequence due to natural variation or mutagenesis, as described in detail in subsection I above. Accordingly, in another embodiment, the MR protein is a protein which comprises an amino acid sequence which is at least about 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, or 60%, preferably at least about 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, or 70%, more preferably at least about 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, or 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, or 90%, or 91%, 92%, 93%, 94%, and even more preferably at least about 95%, 96%, 97%, 98%, 99% or more homologous to an entire amino acid sequence of Appendix B and which has at least one of the MR activities described herein. Ranges and identity values intermediate to the above-recited values, (e.g., 70-90% identical or 80-95% identical) are also intended to be encompassed by the present invention. For example, ranges of identity values using a combination of any of the above values recited as upper and/or lower limits are intended to be included. In another embodiment, the invention pertains to a full length C. glutamicum protein which is substantially homologous to an entire amino acid sequence of Appendix B.
  • Biologically active portions of an MR protein include peptides comprising amino acid sequences derived from the amino acid sequence of an MR protein, e.g., the an amino acid sequence shown in Appendix B or the amino acid sequence of a protein homologous to an MR protein, which include fewer amino acids than a full length MR protein or the full length protein which is homologous to an MR protein, and exhibit at least one activity of an MR protein. Typically, biologically active portions (peptides, e.g., peptides which are, for example, 5, 10, 15, 20, 30, 35, 36, 37, 38, 39, 40, 50, 100 or more amino acids in length) comprise a domain or motif with at least one activity of an MR protein. Moreover, other biologically active portions, in which other regions of the protein are deleted, can be prepared by recombinant techniques and evaluated for one or more of the activities described herein. Preferably, the biologically active portions of an MR protein include one or more selected domains/motifs or portions thereof having biological activity.
  • MR proteins are preferably produced by recombinant DNA techniques. For example, a nucleic acid molecule encoding the protein is cloned into an expression vector (as described above), the expression vector is introduced into a host cell (as described above) and the MR protein is expressed in the host cell. The MR protein can then be isolated from the cells by an appropriate purification scheme using standard protein purification techniques. Alternative to recombinant expression, an MR protein, polypeptide, or peptide can be synthesized chemically using standard peptide synthesis techniques. Moreover, native MR protein can be isolated from cells (e.g., endothelial cells), for example using an anti-MR antibody, which can be produced by standard techniques utilizing an MR protein or fragment thereof of this invention.
  • The invention also provides MR chimeric or fusion proteins. As used herein, an MR “chimeric protein” or “fusion protein” comprises an MR polypeptide operatively linked to a non-MR polypeptide. An “MR polypeptide” refers to a polypeptide having an amino acid sequence corresponding to an MR protein, whereas a “non-MR polypeptide” refers to a polypeptide having an amino acid sequence corresponding to a protein which is not substantially homologous to the MR protein, e.g., a protein which is different from the MR protein and which is derived from the same or a different organism. Within the fusion protein, the term “operatively linked” is intended to indicate that the MR polypeptide and the non-MR polypeptide are fused in-frame to each other. The non-MR polypeptide can be fused to the N-terminus or C-terminus of the MR polypeptide. For example, in one embodiment the fusion protein is a GST-MR fusion protein in which the MR sequences are fused to the C-terminus of the GST sequences. Such fusion proteins can facilitate the purification of recombinant MR proteins. In another embodiment, the fusion protein is an MR protein containing a heterologous signal sequence at its N-terminus. In certain host cells (e.g., mammalian host cells), expression and/or secretion of an MR protein can be increased through use of a heterologous signal sequence.
  • Preferably, an MR chimeric or fusion protein of the invention is produced by standard recombinant DNA techniques. For example, DNA fragments coding for the different polypeptide sequences are ligated together in-frame in accordance with conventional techniques, for example by employing blunt-ended or stagger-ended termini for ligation, restriction enzyme digestion to provide for appropriate termini, filling-in of cohesive ends as appropriate, alkaline phosphatase treatment to avoid undesirable joining, and enzymatic ligation. In another embodiment, the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers. Alternatively, PCR amplification of gene fragments can be carried out using anchor primers which give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed and reamplified to generate a chimeric gene sequence (see, for example, Current Protocols in Molecular Biology, eds. Ausubel et al. John Wiley & Sons: 1992). Moreover, many expression vectors are commercially available that already encode a fusion moiety (e.g., a GST polypeptide). An MR-encoding nucleic acid can be cloned into such an expression vector such that the fusion moiety is linked in-frame to the MR protein.
  • Homologues of the MR protein can be generated by mutagenesis, e.g., discrete point mutation or truncation of the MR protein. As used herein, the term “homologue” refers to a variant form of the MR protein which acts as an agonist or antagonist of the activity of the MR protein. An agonist of the MR protein can retain substantially the same, or a subset, of the biological activities of the MR protein. An antagonist of the MR protein can inhibit one or more of the activities of the naturally occurring form of the MR protein, by, for example, competitively binding to a downstream or upstream member of the MR regulatory cascade which includes the MR protein. Thus, the C. glutamicum MR protein and homologues thereof of the present invention may modulate the activity of one or more metabolic pathways which MR proteins regulate in this microorganism.
  • In an alternative embodiment, homologues of the MR protein can be identified by screening combinatorial libraries of mutants, e.g., truncation mutants, of the MR protein for MR protein agonist or antagonist activity. In one embodiment, a variegated library of MR variants is generated by combinatorial mutagenesis at the nucleic acid level and is encoded by a variegated gene library. A variegated library of MR variants can be produced by, for example, enzymatically ligating a mixture of synthetic oligonucleotides into gene sequences such that a degenerate set of potential MR sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g., for phage display) containing the set of MR sequences therein. There are a variety of methods which can be used to produce libraries of potential MR homologues from a degenerate oligonucleotide sequence. Chemical synthesis of a degenerate gene sequence can be performed in an automatic DNA synthesizer, and the synthetic gene then ligated into an appropriate expression vector. Use of a degenerate set of genes allows for the provision, in one mixture, of all of the sequences encoding the desired set of potential MR sequences. Methods for synthesizing degenerate oligonucleotides are known in the art (see, e.g., Narang, S. A. (1983) Tetrahedron 39:3; Itakura et al. (1984) Annu. Rev. Biochem. 53:323; Itakura et al. (1984) Science 198:1056; Ike et al. (1983) Nucleic Acid Res. 11:477.
  • In addition, libraries of fragments of the MR protein coding can be used to generate a variegated population of MR fragments for screening and subsequent selection of homologues of an MR protein. In one embodiment, a library of coding sequence fragments can be generated by treating a double stranded PCR fragment of an MR coding sequence with a nuclease under conditions wherein nicking occurs only about once per molecule, denaturing the double stranded DNA, renaturing the DNA to form double stranded DNA which can include sense/antisense pairs from different nicked products, removing single stranded portions from reformed duplexes by treatment with S1 nuclease, and ligating the resulting fragment library into an expression vector. By this method, an expression library can be derived which encodes N-terminal, C-terminal and internal fragments of various sizes of the MR protein.
  • Several techniques are known in the art for screening gene products of combinatorial libraries made by point mutations or truncation, and for screening cDNA libraries for gene products having a selected property. Such techniques are adaptable for rapid screening of the gene libraries generated by the combinatorial mutagenesis of MR homologues. The most widely used techniques, which are amenable to high through-put analysis, for screening large gene libraries typically include cloning the gene library into replicable expression vectors, transforming appropriate cells with the resulting library of vectors, and expressing the combinatorial genes under conditions in which detection of a desired activity facilitates isolation of the vector encoding the gene whose product was detected. Recursive ensemble mutagenesis (REM), a new technique which enhances the frequency of functional mutants in the libraries, can be used in combination with the screening assays to identify MR homologues (Arkin and Yourvan (1992) PNAS 89:7811-7815; Delgrave et al. (1993) Protein Engineering 6(3):327-331).
  • In another embodiment, cell based assays can be exploited to analyze a variegated MR library, using methods well known in the art.
  • D. Uses and Methods of the Invention
  • The nucleic acid molecules, proteins, protein homologues, fusion proteins, primers, vectors, and host cells described herein can be used in one or more of the following methods: identification of C. glutamicum and related organisms; mapping of genomes of organisms related to C. glutamicum; identification and localization of C. glutamicum sequences of interest; evolutionary studies; determination of MR protein regions required for function; modulation of an MR protein activity; modulation of the activity of one or more metabolic pathways; and modulation of cellular production of a desired compound, such as a fine chemical.
  • The MR nucleic acid molecules of the invention have a variety of uses. First, they may be used to identify an organism as being Corynebacterium glutamicum or a close relative thereof. Also, they may be used to identify the presence of C. glutamicum or a relative thereof in a mixed population of microorganisms. The invention provides the nucleic acid sequences of a number of C. glutamicum genes; by probing the extracted genomic DNA of a culture of a unique or mixed population of microorganisms under stringent conditions with a probe spanning a region of a C. glutamicum gene which is unique to this organism, one can ascertain whether this organism is present.
  • Although Corynebacterium glutamicum itself is nonpathogenic, it is related to pathogenic species, such as Corynebacterium diphtheriae. Corynebacterium diphtheriae is the causative agent of diphtheria, a rapidly developing, acute, febrile infection which involves both local and systemic pathology. In this disease, a local lesion develops in the upper respiratory tract and involves necrotic injury to epithelial cells; the bacilli secrete toxin which is disseminated through this lesion to distal susceptible tissues of the body. Degenerative changes brought about by the inhibition of protein synthesis in these tissues, which include heart, muscle, peripheral nerves, adrenals, kidneys, liver and spleen, result in the systemic pathology of the disease. Diphtheria continues to have high incidence in many parts of the world, including Africa, Asia, Eastern Europe and the independent states of the former Soviet Union. An ongoing epidemic of diphtheria in the latter two regions has resulted in at least 5,000 deaths since 1990.
  • In one embodiment, the invention provides a method of identifying the presence or activity of Cornyebacterium diphtheriae in a subject. This method includes detection of one or more of the nucleic acid or amino acid sequences of the invention (e.g., the sequences set forth in Appendix A or Appendix B) in a subject, thereby detecting the presence or activity of Corynebacterium diphtheriae in the subject. C. glutamicum and C. diphtheriae are related bacteria, and many of the nucleic acid and protein molecules in C. glutamicum are homologous to C. diphtheriae nucleic acid and protein molecules, and can therefore be used to detect C. diphtheriae in a subject.
  • The nucleic acid and protein molecules of the invention may also serve as markers for specific regions of the genome. This has utility not only in the mapping of the genome, but also for functional studies of C. glutamicum proteins. For example, to identify the region of the genome to which a particular C. glutamicum DNA-binding protein binds, the C. glutamicum genome could be digested, and the fragments incubated with the DNA-binding protein. Those which bind the protein may be additionally probed with the nucleic acid molecules of the invention, preferably with readily detectable labels; binding of such a nucleic acid molecule to the genome fragment enables the localization of the fragment to the genome map of C. glutamicum, and, when performed multiple times with different enzymes, facilitates a rapid determination of the nucleic acid sequence to which the protein binds. Further, the nucleic acid molecules of the invention may be sufficiently homologous to the sequences of related species such that these nucleic acid molecules may serve as markers for the construction of a genomic map in related bacteria, such as Brevibacterium lactofermentum.
  • The MR nucleic acid molecules of the invention are also useful for evolutionary and protein structural studies. The metabolic processes in which the molecules of the invention participate are utilized by a wide variety of prokaryotic and eukaryotic cells; by comparing the sequences of the nucleic acid molecules of the present invention to those encoding similar enzymes from other organisms, the evolutionary relatedness of the organisms can be assessed. Similarly, such a comparison permits an assessment of which regions of the sequence are conserved and which are not, which may aid in determining those regions of the protein which are essential for the functioning of the enzyme. This type of determination is of value for protein engineering studies and may give an indication of what the protein can tolerate in terms of mutagenesis without losing function.
  • Manipulation of the MR nucleic acid molecules of the invention may result in the production of MR proteins having functional differences from the wild-type MR proteins. These proteins may be improved in efficiency or activity, may be present in greater numbers in the cell than is usual, or may be decreased in efficiency or activity.
  • The invention provides methods for screening molecules which modulate the activity of an MR protein, either by interacting with the protein itself or a substrate or binding partner of the MR protein, or by modulating the transcription or translation of an MR nucleic acid molecule of the invention. In such methods, a microorganism expressing one or more MR proteins of the invention is contacted with one or more test compounds, and the effect of each test compound on the activity or level of expression of the MR protein is assessed.
  • Such changes in activity may directly modulate the yield, production, and/or efficiency of production of one or more fine chemicals from C. glutamicum. For example, by optimizing the activity of an MR protein which activates the transcription or translation of a gene encoding a biosynthetic protein for a desired fine chemical, or by impairing or abrogating the activity of an MR protein which represses the transcription or translation of such a gene, one may also increase the activity or rate of activity of that biosynthetic pathway due to the presence of increased levels of what may have been a limiting enzyme. Similarly, by altering the activity of an MR protein such that it constitutively posttranslationally inactivates a protein involved in a degradation pathway for a desired fine chemical, or by altering the activity of an MR protein such that it constitutively represses the transcription or translation of such a gene, one may increase the yield and/or rate of production of the fine chemical from the cell, due to decreased degradation of the compound.
  • Further, by modulating the activity of one or more MR proteins, one may indirectly stimulate the production or improve the rate of production of one or more fine chemicals from the cell due to the interrelatedness of disparate metabolic pathways. For example, by increasing the yield, production, and/or efficiency of production by activating the expression of one or more lysine biosynthetic enzymes, one may concomitantly increase the expression of other compounds, such as other amino acids, which the cell would naturally require in greater quantities when lysine is required in greater quantities. Also, regulation of metabolism throughout the cell may be altered such that the cell is better able to grow or replicate under the environmental conditions of fermentative culture (where nutrient and oxygen supplies may be poor and possibly toxic waste products in the environment may be at high levels). For example, by mutagenizing an MR protein which represses the synthesis of molecules necessary for cell membrane production in response to high levels of waste products in the extracellular medium (in order to block cell growth and division in suboptimal growth conditions) such that it no longer is able to repress such synthesis, one may increase the growth and multiplication of the cell in cultures even when the growth conditions are suboptimal. Such enhanced growth or viability should also increase the yields and/or rate of production of a desired fine chemical from fermentative culture, due to the relatively greater number of cells producing this compound in the culture.
  • The aforementioned mutagenesis strategies for MR proteins to result in increased yields of a fine chemical from C. glutamicum are not meant to be limiting; variations on these strategies will be readily apparent to one of ordinary skill in the art. Using such strategies, and incorporating the mechanisms disclosed herein, the nucleic acid and protein molecules of the invention may be utilized to generate C. glutamicum or related strains of bacteria expressing mutated MR nucleic acid and protein molecules such that the yield and/or efficiency of production of a desired compound is improved. This desired compound may be any natural product of C. glutamicum, which includes the final products of biosynthesis pathways and intermediates of naturally-occurring metabolic pathways, as well as molecules which do not naturally occur in the metabolism of C. glutamicum, but which are produced by a C. glutamicum strain of the invention.
  • This invention is further illustrated by the following examples which should not be construed as limiting. The contents of all references, patent applications, patents, published patent applications, Tables, Appendices, and the sequence listing cited throughout this application are hereby incorporated by reference.
  • EXEMPLIFICATION Example 1 Preparation of Total Genomic DNA of Corynebacterium glutamicum ATCC 13032
  • A culture of Corynebacterium glutamicum (ATCC 13032) was grown overnight at 30° C. with vigorous shaking in BHI medium (Difco). The cells were harvested by centrifugation, the supernatant was discarded and the cells were resuspended in 5 ml buffer-I (5% of the original volume of the culture—all indicated volumes have been calculated for 100 ml of culture volume). Composition of buffer-I: 140.34 g/l sucrose, 2.46 g/l MgSO4×7H2O, 10 ml/l KH2PO4 solution (100 g/l, adjusted to pH 6.7 with KOH), 50 ml/l M12 concentrate (10 g/l (NH4)2SO4, 1 g/l NaCl, 2 μl MgSO4×7H2O, 0.2 g/l CaCl2, 0.5 g/l yeast extract (Difco), 10 ml/l trace-elements-mix (200 mg/l FeSO4×H2O, 10 mg/l ZnSO4×7H2O, 3 mg/l MnCl2×4H2O, 30 mg/l H3BO3 20 mg/l CoCl2×6H2O, 1 mg/l NiCl2×6H2O, 3 mg/l Na2MoO4×2H2O, 500 mg/l complexing agent (EDTA or critic acid), 100 ml/l vitamins-mix (0.2 mg/l biotin, 0.2 mg/l folic acid, 20 mg/l p-amino benzoic acid, 20 mg/l riboflavin, 40 mg/l ca-panthothenate, 140 mg/l nicotinic acid, 40 mg/l pyridoxole hydrochloride, 200 mg/l myo-inositol). Lysozyme was added to the suspension to a final concentration of 2.5 mg/ml. After an approximately 4 h incubation at 37° C., the cell wall was degraded and the resulting protoplasts are harvested by centrifugation. The pellet was washed once with 5 ml buffer-I and once with 5 ml TE-buffer (10 mM Tris-HCl, 1 mM EDTA, pH 8). The pellet was resuspended in 4 ml TE-buffer and 0.5 ml SDS solution (10%) and 0.5 ml NaCl solution (5 M) are added. After adding of proteinase K to a final concentration of 200 μg/ml, the suspension is incubated for ca. 18 h at 37° C. The DNA was purified by extraction with phenol, phenol-chloroform-isoamylalcohol and chloroform-isoamylalcohol using standard procedures. Then, the DNA was precipitated by adding 1/50 volume of 3 M sodium acetate and 2 volumes of ethanol, followed by a 30 min incubation at −20° C. and a 30 min centrifugation at 12,000 rpm in a high speed centrifuge using a SS34 rotor (Sorvall). The DNA was dissolved in 1 ml TE-buffer containing 20 μg/ml RNaseA and dialysed at 4° C. against 1000 ml TE-buffer for at least 3 hours. During this time, the buffer was exchanged 3 times. To aliquots of 0.4 ml of the dialysed DNA solution, 0.4 ml of 2 M LiCl and 0.8 ml of ethanol are added. After a 30 min incubation at −20° C., the DNA was collected by centrifugation (13,000 rpm, Biofuge Fresco, Heraeus, Hanau, Germany). The DNA pellet was dissolved in TE-buffer. DNA prepared by this procedure could be used for all purposes, including southern blotting or construction of genomic libraries.
  • Example 2 Construction of Genomic Libraries in Escherichia coli of Corynebacterium glutamicum ATCC13032
  • Using DNA prepared as described in Example 1, cosmid and plasmid libraries were constructed according to known and well established methods (see e.g., Sambrook, J. et al. (1989) “Molecular Cloning: A Laboratory Manual”, Cold Spring Harbor Laboratory Press, or Ausubel, F. M. et al. (1994) “Current Protocols in Molecular Biology”, John Wiley & Sons.)
  • Any plasmid or cosmid could be used. Of particular use were the plasmids pBR322 (Sutcliffe, J. G. (1979) Proc. Natl. Acad. Sci. USA, 75:3737-3741); pACYC177 (Change & Cohen (1978) J. Bacteriol 134:1141-1156), plasmids of the pBS series (pBSSK+, pBSSK− and others; Stratagene, LaJolla, USA), or cosmids as SuperCos1 (Stratagene, LaJolla, USA) or Lorist6 (Gibson, T. J., Rosenthal A. and Waterson, R. H. (1987) Gene 53:283-286. Gene libraries specifically for use in C. glutamicum may be constructed using plasmid pSL109 (Lee, H.-S. and A. J. Sinskey (1994) J. Microbiol. Biotechnol. 4: 256-263).
  • Example 3 DNA Sequencing and Computational Functional Analysis
  • Genomic libraries as described in Example 2 were used for DNA sequencing according to standard methods, in particular by the chain termination method using ABI377 sequencing machines (see e.g., Fleischman, R. D. et al. (1995) “Whole-genome Random Sequencing and Assembly of Haemophilus Influenzae Rd., Science, 269:496-512). Sequencing primers with the following nucleotide sequences were used: 5′-GGAAACAGTATGACCATG-3′ or 5′-GTAAAACGACGGCCAGT-3′.
  • Example 4 In Vivo Mutagenesis
  • In vivo mutagenesis of Corynebacterium glutamicum can be performed by passage of plasmid (or other vector) DNA through E. coli or other microorganisms (e.g. Bacillus spp. or yeasts such as Saccharomyces cerevisiae) which are impaired in their capabilities to maintain the integrity of their genetic information. Typical mutator strains have mutations in the genes for the DNA repair system (e.g., mutHLS, mutD, mutT, etc.; for reference, see Rupp, W. D. (1996) DNA repair mechanisms, in: Escherichia coli and Salmonella, p. 2277-2294, ASM: Washington.) Such strains are well known to one of ordinary skill in the art. The use of such strains is illustrated, for example, in Greener, A. and Callahan, M. (1994) Strategies 7: 32-34.
  • Example 5 DNA Transfer Between Escherichia coli and Corynebacterium glutamicum
  • Several Corynebacterium and Brevibacterium species contain endogenous plasmids (as e.g., pHM1519 or pBL1) which replicate autonomously (for review see, e.g., Martin, J. F. et al. (1987) Biotechnology, 5:137-146). Shuttle vectors for Escherichia coli and Corynebacterium glutamicum can be readily constructed by using standard vectors for E. coli (Sambrook, J. et al. (1989), “Molecular Cloning: A Laboratory Manual”, Cold Spring Harbor Laboratory Press or Ausubel, F. M. et al. (1994) “Current Protocols in Molecular Biology”, John Wiley & Sons) to which a origin or replication for and a suitable marker from Corynebacterium glutamicum is added. Such origins of replication are preferably taken from endogenous plasmids isolated from Corynebacterium and Brevibacterium species. Of particular use as transformation markers for these species are genes for kanamycin resistance (such as those derived from the Tn5 or Tn903 transposons) or chloramphenicol (Winnacker, E. L. (1987) “From Genes to Clones—Introduction to Gene Technology, VCH, Weinheim). There are numerous examples in the literature of the construction of a wide variety of shuttle vectors which replicate in both E. coli and C. glutamicum, and which can be used for several purposes, including gene over-expression (for reference, see e.g., Yoshihama, M. et al. (1985) J. Bacteriol. 162:591-597, Martin J. F. et al. (1987) Biotechnology, 5:137-146 and Eikmanns, B. J. et al. (1991) Gene, 102:93-98).
  • Using standard methods, it is possible to clone a gene of interest into one of the shuttle vectors described above and to introduce such a hybrid vectors into strains of Corynebacterium glutamicum. Transformation of C. glutamicum can be achieved by protoplast transformation (Kastsumata, R. et al. (1984) J. Bacteriol. 159306-311), electroporation (Liebl, E. et al. (1989) FEMS Microbiol. Letters, 53:399-303) and in cases where special vectors are used, also by conjugation (as described e.g. in Schäfer, A et al. (1990) J. Bacteriol. 172:1663-1666). It is also possible to transfer the shuttle vectors for C. glutamicum to E. coli by preparing plasmid DNA from C. glutamicum (using standard methods well-known in the art) and transforming it into E. coli. This transformation step can be performed using standard methods, but it is advantageous to use an Mcr-deficient E. coli strain, such as NM522 (Gough & Murray (1983) J. Mol. Biol. 166:1-19).
  • Genes may be overexpressed in C. glutamicum strains using plasmids which comprise pCG1 (U.S. Pat. No. 4,617,267) or fragments thereof, and optionally the gene for kanamycin resistance from TN903 (Grindley, N. D. and Joyce, C. M. (1980) Proc. Natl. Acad. Sci. USA 77(12): 7176-7180). In addition, genes may be overexpressed in C. glutamicum strains using plasmid pSL109 (Lee, H.-S. and A. J. Sinskey (1994) J. Microbiol. Biotechnol. 4: 256-263).
  • Aside from the use of replicative plasmids, gene overexpression can also be achieved by integration into the genome. Genomic integration in C. glutamicum or other Corynebacterium or Brevibacterium species may be accomplished by well-known methods, such as homologous recombination with genomic region(s), restriction endonuclease mediated integration (REMI) (see, e.g., DE Patent 19823834), or through the use of transposons. It is also possible to modulate the activity of a gene of interest by modifying the regulatory regions (e.g., a promoter, a repressor, and/or an enhancer) by sequence modification, insertion, or deletion using site-directed methods (such as homologous recombination) or methods based on random events (such as transposon mutagenesis or REMI). Nucleic acid sequences which function as transcriptional terminators may also be inserted 3′ to the coding region of one or more genes of the invention; such terminators are well-known in the art and are described, for example, in Winnacker, E. L. (1987) From Genes to Clones—Introduction to Gene Technology. VCH: Weinheim.
  • Example 6 Assessment of the Expression of the Mutant Protein
  • Observations of the activity of a mutated protein in a transformed host cell rely on the fact that the mutant protein is expressed in a similar fashion and in a similar quantity to that of the wild-type protein. A useful method to ascertain the level of transcription of the mutant gene (an indicator of the amount of mRNA available for translation to the gene product) is to perform a Northern blot (for reference see, for example, Ausubel et al. (1988) Current Protocols in Molecular Biology, Wiley: New York), in which a primer designed to bind to the gene of interest is labeled with a detectable tag (usually radioactive or chemiluminescent), such that when the total RNA of a culture of the organism is extracted, run on gel, transferred to a stable matrix and incubated with this probe, the binding and quantity of binding of the probe indicates the presence and also the quantity of mRNA for this gene. This information is evidence of the degree of transcription of the mutant gene. Total cellular RNA can be prepared from Corynebacterium glutamicum by several methods, all well-known in the art, such as that described in Bormann, E. R. et al. (1992) Mol. Microbiol. 6: 317-326.
  • To assess the presence or relative quantity of protein translated from this mRNA, standard techniques, such as a Western blot, may be employed (see, for example, Ausubel et al. (1988) Current Protocols in Molecular Biology, Wiley: New York). In this process, total cellular proteins are extracted, separated by gel electrophoresis, transferred to a matrix such as nitrocellulose, and incubated with a probe, such as an antibody, which specifically binds to the desired protein. This probe is generally tagged with a chemiluminescent or colorimetric label which may be readily detected. The presence and quantity of label observed indicates the presence and quantity of the desired mutant protein present in the cell.
  • Example 7 Growth of Genetically Modified Corynebacterium glutamicum—Media and Culture Conditions
  • Genetically modified Corynebacteria are cultured in synthetic or natural growth media. A number of different growth media for Corynebacteria are both well-known and readily available (Lieb et al. (1989) Appl. Microbiol. Biotechnol., 32:205-210; von der Osten et al. (1998) Biotechnology Letters, 11:11-16; Patent DE 4,120,867; Liebl (1992) “The Genus Corynebacterium, in: The Procaryotes, Volume II, Balows, A. et al., eds. Springer-Verlag). These media consist of one or more carbon sources, nitrogen sources, inorganic salts, vitamins and trace elements. Preferred carbon sources are sugars, such as mono-, di-, or polysaccharides. For example, glucose, fructose, mannose, galactose, ribose, sorbose, ribulose, lactose, maltose, sucrose, raffinose, starch or cellulose serve as very good carbon sources. It is also possible to supply sugar to the media via complex compounds such as molasses or other by-products from sugar refinement. It can also be advantageous to supply mixtures of different carbon sources. Other possible carbon sources are alcohols and organic acids, such as methanol, ethanol, acetic acid or lactic acid. Nitrogen sources are usually organic or inorganic nitrogen compounds, or materials which contain these compounds. Exemplary nitrogen sources include ammonia gas or ammonia salts, such as NH4Cl or (NH4)2SO4, NH4OH, nitrates, urea, amino acids or complex nitrogen sources like corn steep liquor, soy bean flour, soy bean protein, yeast extract, meat extract and others.
  • Inorganic salt compounds which may be included in the media include the chloride-, phosphorous- or sulfate-salts of calcium, magnesium, sodium, cobalt, molybdenum, potassium, manganese, zinc, copper and iron. Chelating compounds can be added to the medium to keep the metal ions in solution. Particularly useful chelating compounds include dihydroxyphenols, like catechol or protocatechuate, or organic acids, such as citric acid. It is typical for the media to also contain other growth factors, such as vitamins or growth promoters, examples of which include biotin, riboflavin, thiamin, folic acid, nicotinic acid, pantothenate and pyridoxin. Growth factors and salts frequently originate from complex media components such as yeast extract, molasses, corn steep liquor and others. The exact composition of the media compounds depends strongly on the immediate experiment and is individually decided for each specific case. Information about media optimization is available in the textbook “Applied Microbiol. Physiology, A Practical Approach (eds. P. M. Rhodes, P. F. Stanbury, IRL Press (1997) pp. 53-73, ISBN 0 19 963577 3). It is also possible to select growth media from commercial suppliers, like standard 1 (Merck) or BHI (grain heart infusion, DIFCO) or others.
  • All medium components are sterilized, either by heat (20 minutes at 1.5 bar and 121° C.) or by sterile filtration. The components can either be sterilized together or, if necessary, separately. All media components can be present at the beginning of growth, or they can optionally be added continuously or batchwise.
  • Culture conditions are defined separately for each experiment. The temperature should be in a range between 15° C. and 45° C. The temperature can be kept constant or can be altered during the experiment. The pH of the medium should be in the range of 5 to 8.5, preferably around 7.0, and can be maintained by the addition of buffers to the media. An exemplary buffer for this purpose is a potassium phosphate buffer. Synthetic buffers such as MOPS, HEPES, ACES and others can alternatively or simultaneously be used. It is also possible to maintain a constant culture pH through the addition of NaOH or NH4OH during growth. If complex medium components such as yeast extract are utilized, the necessity for additional buffers may be reduced, due to the fact that many complex compounds have high buffer capacities. If a fermentor is utilized for culturing the micro-organisms, the pH can also be controlled using gaseous ammonia.
  • The incubation time is usually in a range from several hours to several days. This time is selected in order to permit the maximal amount of product to accumulate in the broth. The disclosed growth experiments can be carried out in a variety of vessels, such as microtiter plates, glass tubes, glass flasks or glass or metal fermentors of different sizes. For screening a large number of clones, the microorganisms should be cultured in microtiter plates, glass tubes or shake flasks, either with or without baffles. Preferably 100 ml shake flasks are used, filled with 10% (by volume) of the required growth medium. The flasks should be shaken on a rotary shaker (amplitude 25 mm) using a speed-range of 100-300 rpm. Evaporation losses can be diminished by the maintenance of a humid atmosphere; alternatively, a mathematical correction for evaporation losses should be performed.
  • If genetically modified clones are tested, an unmodified control clone or a control clone containing the basic plasmid without any insert should also be tested. The medium is inoculated to an OD600 of O.5-1.5 using cells grown on agar plates, such as CM plates (10 g/l glucose, 2.5 g/l NaCl, 2 g/l urea, 10 g/l polypeptone, 5 g/l yeast extract, 5 g/l meat extract, 22 g/l NaCl, 2 g/l urea, 10 g/l polypeptone, 5 g/l yeast extract, 5 g/l meat extract, 22 g/l agar, pH 6.8 with 2M NaOH) that had been incubated at 30° C. Inoculation of the media is accomplished by either introduction of a saline suspension of C. glutamicum cells from CM plates or addition of a liquid preculture of this bacterium.
  • Example 8 In Vitro Analysis of the Function of Mutant Proteins
  • The determination of activities and kinetic parameters of enzymes is well established in the art. Experiments to determine the activity of any given altered enzyme must be tailored to the specific activity of the wild-type enzyme, which is well within the ability of one of ordinary skill in the art. Overviews about enzymes in general, as well as specific details concerning structure, kinetics, principles, methods, applications and examples for the determination of many enzyme activities may be found, for example, in the following references: Dixon, M., and Webb, E. C., (1979) Enzymes. Longmans: London; Fersht, (1985) Enzyme Structure and Mechanism. Freeman: New York; Walsh, (1979) Enzymatic Reaction Mechanisms. Freeman: San Francisco; Price, N. C., Stevens, L. (1982) Fundamentals of Enzymology. Oxford Univ. Press: Oxford; Boyer, P. D., ed. (1983) The Enzymes, 3rd ed. Academic Press: New York; Bisswanger, H., (1994) Enzymkinetik, 2nd ed. VCH: Weinheim (ISBN 3527300325); Bergmeyer, H. U., Bergmeyer, J., Graβl, M., eds. (1983-1986) Methods of Enzymatic Analysis, 3rd ed., vol. I-XII, Verlag Chemie: Weinheim; and Ullmann's Encyclopedia of Industrial Chemistry (1987) vol. A9, “Enzymes”. VCH: Weinheim, p. 352-363.
  • The activity of proteins which bind to DNA can be measured by several well-established methods, such as DNA band-shift assays (also called gel retardation assays). The effect of such proteins on the expression of other molecules can be measured using reporter gene assays (such as that described in Kolmar, H. et al. (1995) EMBO J. 14: 3895-3904 and references cited therein). Reporter gene test systems are well known and established for applications in both pro- and eukaryotic cells, using enzymes such as beta-galactosidase, green fluorescent protein, and several others.
  • The determination of activity of membrane-transport proteins can be performed according to techniques such as those described in Gennis, R. B. (1989) “Pores, Channels and Transporters”, in Biomembranes, Molecular Structure and Function, Springer: Heidelberg, p. 85-137; 199-234; and 270-322.
  • Example 9 Analysis of Impact of Mutant Protein on the Production of the Desired Product
  • The effect of the genetic modification in C. glutamicum on production of a desired compound (such as an amino acid) can be assessed by growing the modified microorganism under suitable conditions (such as those described above) and analyzing the medium and/or the cellular component for increased production of the desired product (i.e., an amino acid). Such analysis techniques are well known to one of ordinary skill in the art, and include spectroscopy, thin layer chromatography, staining methods of various kinds, enzymatic and microbiological methods, and analytical chromatography such as high performance liquid chromatography (see, for example, Ullman, Encyclopedia of Industrial Chemistry, vol. A2, p. 89-90 and p. 443-613, VCH: Weinheim (1985); Fallon, A. et al., (1987) “Applications of HPLC in Biochemistry” in: Laboratory Techniques in Biochemistry and Molecular Biology, vol. 17; Rehm et al. (1993) Biotechnology, vol. 3, Chapter III: “Product recovery and purification”, page 469-714, VCH: Weinheim; Belter, P. A. et al. (1988) Bioseparations: downstream processing for biotechnology, John Wiley and Sons; Kennedy, J. F. and Cabral, J. M. S. (1992) Recovery processes for biological materials, John Wiley and Sons; Shaeiwitz, J. A. and Henry, J. D. (1988) Biochemical separations, in: Ulmann's Encyclopedia of Industrial Chemistry, vol. B3, Chapter 11, page 1-27, VCH: Weinheim; and Dechow, F. J. (1989) Separation and purification techniques in biotechnology, Noyes Publications.)
  • In addition to the measurement of the final product of fermentation, it is also possible to analyze other components of the metabolic pathways utilized for the production of the desired compound, such as intermediates and side-products, to determine the overall yield, production, and/or efficiency of production of the compound. Analysis methods include measurements of nutrient levels in the medium (e.g., sugars, hydrocarbons, nitrogen sources, phosphate, and other ions), measurements of biomass composition and growth, analysis of the production of common metabolites of biosynthetic pathways, and measurement of gasses produced during fermentation. Standard methods for these measurements are outlined in Applied Microbial Physiology, A Practical Approach, P. M. Rhodes and P. F. Stanbury, eds., IRL Press, p. 103-129; 131-163; and 165-192 (ISBN: 0199635773) and references cited therein.
  • Example 10 Purification of the Desired Product from C. glutamicum Culture
  • Recovery of the desired product from the C. glutamicum cells or supernatant of the above-described culture can be performed by various methods well known in the art. If the desired product is not secreted from the cells, the cells can be harvested from the culture by low-speed centrifugation, the cells can be lysed by standard techniques, such as mechanical force or sonication. The cellular debris is removed by centrifugation, and the supernatant fraction containing the soluble proteins is retained for further purification of the desired compound. If the product is secreted from the C. glutamicum cells, then the cells are removed from the culture by low-speed centrifugation, and the supernate fraction is retained for further purification.
  • The supernatant fraction from either purification method is subjected to chromatography with a suitable resin, in which the desired molecule is either retained on a chromatography resin while many of the impurities in the sample are not, or where the impurities are retained by the resin while the sample is not. Such chromatography steps may be repeated as necessary, using the same or different chromatography resins. One of ordinary skill in the art would be well-versed in the selection of appropriate chromatography resins and in their most efficacious application for a particular molecule to be purified. The purified product may be concentrated by filtration or ultrafiltration, and stored at a temperature at which the stability of the product is maximized.
  • There are a wide array of purification methods known to the art and the preceding method of purification is not meant to be limiting. Such purification techniques are described, for example, in Bailey, J. E. & Ollis, D. F. Biochemical Engineering Fundamentals, McGraw-Hill: New York (1986).
  • The identity and purity of the isolated compounds may be assessed by techniques standard in the art. These include high-performance liquid chromatography (HPLC), spectroscopic methods, staining methods, thin layer chromatography, NIRS, enzymatic assay, or microbiologically. Such analysis methods are reviewed in: Patek et al. (1994) Appl. Environ. Microbiol. 60: 133-140; Malakhova et al. (1996) Biotekhnologiya 11: 27-32; and Schmidt et al. (1998) Bioprocess Engineer. 19: 67-70. Ulmann's Encyclopedia of Industrial Chemistry, (1996) vol. A27, VCH: Weinheim, p. 89-90, p. 521-540, p. 540-547, p. 559-566, 575-581 and p. 581-587; Michal, G. (1999) Biochemical Pathways: An Atlas of Biochemistry and Molecular Biology, John Wiley and Sons; Fallon, A. et al. (1987) Applications of HPLC in Biochemistry in: Laboratory Techniques in Biochemistry and Molecular Biology, vol. 17.
  • Example 11 Analysis of the Gene Sequences of the Invention
  • The comparison of sequences and determination of percent homology between two sequences are art-known techniques, and can be accomplished using a mathematical algorithm, such as the algorithm of Karlin and Altschul (1990) Proc. Natl. Acad. Sci. USA 87:2264-68, modified as in Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90:5873-77. Such an algorithm is incorporated into the NBLAST and XBLAST programs (version 2.0) of Altschul, et al. (1990) J. Mol. Biol. 215:403-10. BLAST nucleotide searches can be performed with the NBLAST program, score=100, wordlength=12 to obtain nucleotide sequences homologous to MR nucleic acid molecules of the invention. BLAST protein searches can be performed with the XBLAST program, score=50, wordlength=3 to obtain amino acid sequences homologous to MR protein molecules of the invention. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et al., (1997) Nucleic Acids Res. 25(17):3389-3402. When utilizing BLAST and Gapped BLAST programs, one of ordinary skill in the art will know how to optimize the parameters of the program (e.g., XBLAST and NBLAST) for the specific sequence being analyzed.
  • Another example of a mathematical algorithm utilized for the comparison of sequences is the algorithm of Meyers and Miller ((1988) Comput. Appl. Biosci. 4: 11-17). Such an algorithm is incorporated into the ALIGN program (version 2.0) which is part of the GCG sequence alignment software package. When utilizing the ALIGN program for comparing amino acid sequences, a PAM120 weight residue table, a gap length penalty of 12, and a gap penalty of 4 can be used. Additional algorithms for sequence analysis are known in the art, and include ADVANCE and ADAM. described in Torelli and Robotti (1994) Comput. Appl. Biosci. 10:3-5; and FASTA, described in Pearson and Lipman (1988) P.N.A.S. 85:2444-8.
  • The percent homology between two amino acid sequences can also be accomplished using the GAP program in the GCG software package (available at http://www.gcg.com), using either a Blosum 62 matrix or a PAM250 matrix, and a gap weight of 12, 10, 8, 6, or 4 and a length weight of 2, 3, or 4. The percent homology between two nucleic acid sequences can be accomplished using the GAP program in the GCG software package, using standard parameters, such as a gap weight of 50 and a length weight of 3.
  • A comparative analysis of the gene sequences of the invention with those present in Genbank has been performed using techniques known in the art (see, e.g., Bexevanis and Ouellette, eds. (1998) Bioinformatics: A Practical Guide to the Analysis of Genes and Proteins. John Wiley and Sons: New York). The gene sequences of the invention were compared to genes present in Genbank in a three-step process. In a first step, a BLASTN analysis (e.g., a local alignment analysis) was performed for each of the sequences of the invention against the nucleotide sequences present in Genbank, and the top 500 hits were retained for further analysis. A subsequent FASTA search (e.g., a combined local and global alignment analysis, in which limited regions of the sequences are aligned) was performed on these 500 hits. Each gene sequence of the invention was subsequently globally aligned to each of the top three FASTA hits, using the GAP program in the GCG software package (using standard parameters). In order to obtain correct results, the length of the sequences extracted from Genbank were adjusted to the length of the query sequences by methods well-known in the art. The results of this analysis are set forth in Table 4. The resulting data is identical to that which would have been obtained had a GAP (global) analysis alone been performed on each of the genes of the invention in comparison with each of the references in Genbank, but required significantly reduced computational time as compared to such a database-wide GAP (global) analysis. Sequences of the invention for which no alignments above the cutoff values were obtained are indicated on Table 4 by the absence of alignment information. It will further be understood by one of ordinary skill in the art that the GAP alignment homology percentages set forth in Table 4 under the heading “% homology (GAP)” are listed in the European numerical format, wherein a ‘,’ represents a decimal point. For example, a value of “40,345” in this column represents “40.345%”.
  • Example 12 Construction and Operation of DNA Microarrays
  • The sequences of the invention may additionally be used in the construction and application of DNA microarrays (the design, methodology, and uses of DNA arrays are well known in the art, and are described, for example, in Schena, M. et al. (1995) Science 270: 467-470; Wodicka, L. et al. (1997) Nature Biotechnology 15: 1359-1367; DeSaizieu, A. et al. (1998) Nature Biotechnology 16: 45-48; and DeRisi, J. L. et al. (1997) Science 278: 680-686).
  • DNA microarrays are solid or flexible supports consisting of nitrocellulose, nylon, glass, silicone, or other materials. Nucleic acid molecules may be attached to the surface in an ordered manner. After appropriate labeling, other nucleic acids or nucleic acid mixtures can be hybridized to the immobilized nucleic acid molecules, and the label may be used to monitor and measure the individual signal intensities of the hybridized molecules at defined regions. This methodology allows the simultaneous quantification of the relative or absolute amount of all or selected nucleic acids in the applied nucleic acid sample or mixture. DNA microarrays, therefore, permit an analysis of the expression of multiple (as many as 6800 or more) nucleic acids in parallel (see, e.g., Schena, M. (1996) BioEssays 18(5): 427-431).
  • The sequences of the invention may be used to design oligonucleotide primers which are able to amplify defined regions of one or more C. glutamicum genes by a nucleic acid amplification reaction such as the polymerase chain reaction. The choice and design of the 5′ or 3′ oligonucleotide primers or of appropriate linkers allows the covalent attachment of the resulting PCR products to the surface of a support medium described above (and also described, for example, Schena, M. et al. (1995) Science 270: 467-470).
  • Nucleic acid microarrays may also be constructed by in situ oligonucleotide synthesis as described by Wodicka, L. et al. (1997) Nature Biotechnology 15: 1359-1367. By photolithographic methods, precisely defined regions of the matrix are exposed to light. Protective groups which are photolabile are thereby activated and undergo nucleotide addition, whereas regions that are masked from light do not undergo any modification. Subsequent cycles of protection and light activation permit the synthesis of different oligonucleotides at defined positions. Small, defined regions of the genes of the invention may be synthesized on microarrays by solid phase oligonucleotide synthesis.
  • The nucleic acid molecules of the invention present in a sample or mixture of nucleotides may be hybridized to the microarrays. These nucleic acid molecules can be labeled according to standard methods. In brief, nucleic acid molecules (e.g., mRNA molecules or DNA molecules) are labeled by the incorporation of isotopically or fluorescently labeled nucleotides, e.g., during reverse transcription or DNA synthesis. Hybridization of labeled nucleic acids to microarrays is described (e.g., in Schena, M. et al. (1995) supra; Wodicka, L. et al. (1997), supra; and DeSaizieu A. et al. (1998), supra). The detection and quantification of the hybridized molecule are tailored to the specific incorporated label. Radioactive labels can be detected, for example, as described in Schena, M. et al. (1995) supra) and fluorescent labels may be detected, for example, by the method of Shalon et al. (1996) Genome Research 6: 639-645).
  • The application of the sequences of the invention to DNA microarray technology, as described above, permits comparative analyses of different strains of C. glutamicum or other Corynebacteria. For example, studies of inter-strain variations based on individual transcript profiles and the identification of genes that are important for specific and/or desired strain properties such as pathogenicity, productivity and stress tolerance are facilitated by nucleic acid array methodologies. Also, comparisons of the profile of expression of genes of the invention during the course of a fermentation reaction are possible using nucleic acid array technology.
  • Example 13 Analysis of the Dynamics of Cellular Protein Populations (Proteomics)
  • The genes, compositions, and methods of the invention may be applied to study the interactions and dynamics of populations of proteins, termed ‘proteomics’. Protein populations of interest include, but are not limited to, the total protein population of C. glutamicum (e.g., in comparison with the protein populations of other organisms), those proteins which are active under specific environmental or metabolic conditions (e.g., during fermentation, at high or low temperature, or at high or low pH), or those proteins which are active during specific phases of growth and development.
  • Protein populations can be analyzed by various well-known techniques, such as gel electrophoresis. Cellular proteins may be obtained, for example, by lysis or extraction, and may be separated from one another using a variety of electrophoretic techniques. Sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) separates proteins largely on the basis of their molecular weight. Isoelectric focusing polyacrylamide gel electrophoresis (IEF-PAGE) separates proteins by their isoelectric point (which reflects not only the amino acid sequence but also posttranslational modifications of the protein). Another, more preferred method of protein analysis is the consecutive combination of both IEF-PAGE and SDS-PAGE, known as 2-D-gel electrophoresis (described, for example, in Hermann et al. (1998) Electrophoresis 19: 3217-3221; Fountoulakis et al. (1998) Electrophoresis 19: 1193-1202; Langen et al. (1997) Electrophoresis 18: 1184-1192; Antelmann et al. (1997) Electrophoresis 18: 1451-1463). Other separation techniques may also be utilized for protein separation, such as capillary gel electrophoresis; such techniques are well known in the art.
  • Proteins separated by these methodologies can be visualized by standard techniques, such as by staining or labeling. Suitable stains are known in the art, and include Coomassie Brilliant Blue, silver stain, or fluorescent dyes such as Sypro Ruby (Molecular Probes). The inclusion of radioactively labeled amino acids or other protein precursors (e.g., 35S-methionine, 35S-cysteine, 14C-labelled amino acids, 15N-amino acids, 15NO3 or 15NH4 + or 13C-labelled amino acids) in the medium of C. glutamicum permits the labeling of proteins from these cells prior to their separation. Similarly, fluorescent labels may be employed. These labeled proteins can be extracted, isolated and separated according to the previously described techniques.
  • Proteins visualized by these techniques can be further analyzed by measuring the amount of dye or label used. The amount of a given protein can be determined quantitatively using, for example, optical methods and can be compared to the amount of other proteins in the same gel or in other gels. Comparisons of proteins on gels can be made, for example, by optical comparison, by spectroscopy, by image scanning and analysis of gels, or through the use of photographic films and screens. Such techniques are well-known in the art.
  • To determine the identity of any given protein, direct sequencing or other standard techniques may be employed. For example, N- and/or C-terminal amino acid sequencing (such as Edman degradation) may be used, as may mass spectrometry (in particular MALDI or ESI techniques (see, e.g., Langen et al. (1997) Electrophoresis 18: 1184-1192)). The protein sequences provided herein can be used for the identification of C. glutamicum proteins by these techniques.
  • The information obtained by these methods can be used to compare patterns of protein presence, activity, or modification between different samples from various biological conditions (e.g., different organisms, time points of fermentation, media conditions, or different biotopes, among others). Data obtained from such experiments alone, or in combination with other techniques, can be used for various applications, such as to compare the behavior of various organisms in a given (e.g., metabolic) situation, to increase the productivity of strains which produce fine chemicals or to increase the efficiency of the production of fine chemicals.
  • EQUIVALENTS
  • Those of ordinary skill in the art will recognize, or will be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.
    TABLE 1
    GENES INCLUDED IN THE APPLICATION
    Nucleic Acid
    SEQ ID NO Amino Acid SEQ ID NO Identification Code Contig. NT Start NT Stop Function
    1 2 RXN03181 VV0338 196 609 GLUCOSE-RESISTANCE AMYLASE REGULATOR
    3 4 F RXA02880 GR10018 417 4 TRANSCRIPTIONAL REPRESSOR CYTR
    5 6 RXA00603 GR00159 4982 5434 LEUCINE-RESPONSIVE REGULATORY PROTEIN
    7 8 RXN02946 VV0127 7000 7458 FATTY ACYL RESPONSIVE REGULATOR
    9 10 RXN01845 VV0234 1093 686 FUMARATE AND NITRATE REDUCTION REGULATORY PROTEIN
    11 12 RXN02910 VV0135 30560 29856 TRANSCRIPTIONAL ACTIVATOR PROTEIN LYSR
    13 14 RXN02553 VV0101 3454 4017 CRYPTIC BETA-GLUCOSIDE BGL OPERON ANTITERMINATOR
    15 16 RXS00686 VV0005 30857 30054 ACETATE OPERON REPRESSOR
    17 18 RXS00774 VV0103 22950 22297 PHOSPHATE TRANSPORT SYSTEM REGULATORY PROTEIN
    19 20 RXN02493 VV0007 8481 9719 PHOSPHATE REGULON SENSOR PROTEIN PHOR (EC 2.7.3.—)
    21 22 F RXA02493 GR00720 2931 4169 regulatory gene for the phosphate regulon
    23 24 RXN00631 VV0135 18302 16848 PHOSPHATE REGULON SENSOR PROTEIN PHOR (EC 2.7.3.—)
    Genes for signal transduction pathways, regulation of proteins and transcription
    25 26 RXN00291 VV0041 6431 4860 SENSOR KINASE CITA (EC 2.7.3.—)
    27 28 F RXA00291 GR00047 2 1075 SENSOR KINASE CITA (EC 2.7.3.—)
    29 30 RXA00129 GR00020 6205 4709 SENSOR PROTEIN CPXA (EC 2.7.3.—)
    31 32 RXN00651 VV0109 8052 9383 Hypothetical Sensor Histidine Kinase (EC 2.7.3.—)
    33 34 F RXA00651 GR00169 5450 4119 SENSOR PROTEIN DEGS (EC 2.7.3.—)
    35 36 RXA00006 GR00001 6905 6471 SENSOR PROTEIN FIXL (EC 2.7.3.—)
    37 38 RXA01860 GR00529 2368 1484 SENSOR PROTEIN FIXL (EC 2.7.3.—)
    39 40 RXA01861 GR00529 4332 2368 SENSOR PROTEIN FIXL (EC 2.7.3.—)
    41 42 RXA02669 GR00753 8893 10008 SENSOR PROTEIN RESE (EC 2.7.3.—)
    43 44 RXN01211 VV0169 5106 6362 SENSOR PROTEIN UHPB (EC 2.7.3.—)
    45 46 F RXA01211 GR00349 741 1535 SENSOR PROTEIN UHPB (EC 2.7.3.—)
    47 48 RXA01248 GR00362 165 593 SENSORY TRANSDUCTION PROTEIN REGX3
    49 50 RXA02668 GR00753 8171 8893 SENSORY TRANSDUCTION PROTEIN REGX3
    51 52 RXA02632 GR00748 4863 4168 putative two-component response regulator [Mycobacterium tuberculosis]
    53 54 RXA02631 GR00748 4096 2732 putative two-component sensor [Mycobacterium tuberculosis]
    55 56 RXA00609 GR00161 226 891 TWO COMPONENT RESPONSE REGULATOR
    57 58 RXA00284 GR00045 1318 2382 ANKYRIN HOMOLOG PRECURSOR
    59 60 RXA01827 GR00516 6308 4902 PROTEIN KINASE PKNA
    61 62 RXA00813 GR00219 1345 2475 SECRETORY PROTEIN KINASE
    63 64 RXA01826 GR00516 4902 2965 PUTATIVE SERINE/THREONINE-PROTEIN KINASE PKNB (EC 2.7.1.—)
    65 66 RXA02699 GR00757 1357 3504 PUTATIVE SERINE/THREONINE-PROTEIN KINASE PKNB (EC 2.7.1.—)
    67 68 RXA00319 GR00056 505 80 LOW MOLECULAR WEIGHT PHOSPHOTYROSINE PROTEIN PHOSPHATASE (EC 3.1.3.48)
    69 70 RXA01272 GR00367 25049 24447 PROBABLE LOW MOLECULAR WEIGHT PROTEIN-TYROSINE-PHOSPHATASE EPSP (EC 3.1.3.4
    Figure US20070015252A1-20070118-P00899
    71 72 RXA01830 GR00516 10410 9058 PUTATIVE PHOSPHOPROTEIN PHOSPHATASE
    73 74 RXA02747 GR00764 277 2352 [PROTEIN-PII] URIDYLYLTRANSFERASE (EC 2.7.7.59)
    75 76 RXA02210 GR00648 1922 2485 Hypothetical Transcriptional Regulator
    77 78 RXA00221 GR00032 20855 21073 Hypothetical Transcriptional Regulator
    79 80 RXN00551 VV0079 30941 30471 Hypothetical Transcriptional Regulator
    81 82 F RXA00551 GR00144 352 5 Hypothetical Transcriptional Regulator
    83 84 RXA01763 GR00500 1987 1523 Hypothetical Transcriptional Regulator
    85 86 RXA02667 GR00753 7863 7270 Hypothetical Transcriptional Regulator
    87 88 RXA00348 GR00065 1507 1052 Hypothetical Transcriptional Regulator
    89 90 RXA01500 GR00424 7551 7108 Hypothetical Transcriptional Regulator
    91 92 RXA01125 GR00312 1800 1588 Hypothetical Transcriptional Regulator
    93 94 RXN00822 VV0054 21521 20841 Hypothetical Transcriptional Regulator
    95 96 F RXA00822 GR00221 3073 2393 putative transcriptional regulator
    97 98 RXN00849 VV0067 4701 4381 Hypothetical Transcriptional Regulator
    99 100 F RXA00849 GR00231 378 698 possible transcriptional regulator
    101 102 RXA02698 GR00757 1143 775 PUTATIVE TRANSCRIPTIONAL REGULATOR
    103 104 RXA00350 GR00066 1144 1470 Hypothetical Transcription Inintiation Factor
    105 106 RXA02830 GR00817 3 497 Helix-turn-helix domain-containing transcription regulators
    107 108 RXA00947 GR00259 4164 3829 Helix-turn-helix domain-containing transcriptional regulators
    109 110 RXA01836 GR00517 4370 3666 (AL021287) probable transcriptional regulator [Mycobacterium tuberculosis]
    111 112 RXA00292 GR00047 1078 1731 transcriptional regulator CriR
    113 114 RXA00182 GR00028 4247 7348 POSSIBLE GLOBAL TRANSCRIPTION ACTIVATOR SNF2L
    115 116 RXA02760 GR00767 1154 201 TRANSCRIPTION ANTITERMINATION PROTEIN NUSG
    117 118 RXA02306 GR00663 3214 2924 TRANSCRIPTIONAL REGULATORY PROTEIN CITB
    119 120 RXA00130 GR00020 6985 6308 TRANSCRIPTIONAL REGULATORY PROTEIN CPXR
    121 122 RXA00885 GR00242 11301 12326 HEAT-INDUCIBLE TRANSCRIPTION REPRESSOR HRCA
    123 124 RXA01418 GR00415 776 531 TRANSCRIPTIONAL REPRESSOR SMTB
    125 126 RXA01759 GR00498 4075 4836 TRANSCRIPTIONAL REGULATORY PROTEIN GLTC
    127 128 RXN00363 VV0176 35684 34965 Hypothetical Transcriptional Regulator
    129 130 F RXA00363 GR00073 1929 1246 NTA OPERON TRANSCRIPTIONAL REGULATOR
    131 132 RXA00516 GR00131 592 1311 NTA OPERON TRANSCRIPTIONAL REGULATOR
    133 134 RXA01537 GR00427 4829 4179 NTA OPERON TRANSCRIPTIONAL REGULATOR
    135 136 RXA02494 GR00720 4169 4864 KDP OPERON TRANSCRIPTIONAL REGULATORY PROTEIN KDPE
    137 138 RXA00029 GR00003 8910 8374 PUTATIVE AGA OPERON TRANSCRIPTIONAL REPRESSOR
    139 140 RXA00655 GR00169 9049 8411 putative regulatory protein
    141 142 RXN03136 VV0128 2692 278 Hypothetical Transcriptional Regulator
    143 144 F RXA00645 GR00168 5831 8161 PUTATIVE REGULATORY PROTEIN
    145 146 RXA00593 GR00158 2858 2511 REGULATORY PROTEIN
    147 148 RXA02724 GR00760 870 4 REGULATORY PROTEIN
    149 150 RXA00494 GR00123 768 472 Hypothetical Regulatory Protein
    151 152 RXN01368 VV0091 3096 2785 Hypothetical Regulatory Protein
    153 154 F RXA01368 GR00397 2334 2206 Hypothetical Regulatory Protein
    155 156 RXN00464 VV0086 61883 62656 REGULATORY PROTEIN SIR2 HOMOLOG
    157 158 F RXA00464 GR00117 75 332 REGULATORY PROTEIN SIR2 HOMOLOG
    159 160 RXA01655 GR00460 1458 100 PROBABLE RHIZOPINE CATABOLISM REGULATORY PROTEIN MOCR
    161 162 RXA00126 GR00020 2269 1607 PROBABLE SIGMA(54) MODULATION PROTEIN
    163 164 RXN02450 VV0107 10940 10386 Hypothetical Transcriptional Regulator
    165 166 F RXA02450 GR00710 2533 3087 POTENTIAL ACRAB OPERON REPRESSOR
    167 168 RXA01898 GR00544 1178 1870 OPERON REGULATOR
    169 170 RXA00004 GR00001 4293 3823 NITRILASE REGULATOR
    171 172 RXA01001 GR00284 516 833 hex regulon repressor hexR
    173 174 RXA01375 GR00400 2560 1106 FRNA
    175 176 RXA02831 GR00818 411 4 EXTRAGENIC SUPPRESSOR PROTEIN SUHB
    177 178 RXA01110 GR00306 16399 16971 TETRACYCLINE REPRESSOR PROTEIN CLASS C
    179 180 RXA00253 GR00038 1064 1801 TETRACYCLINE REPRESSOR PROTEIN CLASS E
    181 182 RXA01118 GR00309 1787 2551 regulator of the glyoxylate bypass
    183 184 RXA01840 GR00521 2 655 ALIPHATIC AMIDASE EXPRESSION-REGULATING PROTEIN
    185 186 RXA00400 GR00087 1163 2041 ALS OPERON REGULATORY PROTEIN
    187 188 RXA02787 GR00777 865 2241 ACTIVATOR 1 41 KD SUBUNIT
    189 190 RXA00287 GR00046 1618 1145 ADAPTIVE RESPONSE REGULATORY PROTEIN
    191 192 RXA01687 GR00470 3289 2219 N-ACETYLGLUCOSAMINE REPRESSOR
    193 194 RXA01935 GR00555 8902 7739 N-ACETYLGLUCOSAMINE REPRESSOR
    195 196 RXN02270 VV0020 13880 13260 Hypothetical Transcriptional Regulator
    197 198 F RXA02270 GR00655 5005 4385 member of the regulatory protein family SIR2
    199 200 RXA01241 GR00359 739 1218 LEXA REPRESSOR (EC 3.4.21.88)
    201 202 RXA02127 GR00637 2715 2062 6 ACTVA REGION GENES OF THE ACTINORHODIN BIOSYNTHETIC GENE CLUSTER
    203 204 RXA00583 GR00156 10203 9466 Uncharacterized ACR (translation?)
    205 206 RXA00592 GR00158 2121 1663 Uncharacterized ACR (translation initiation regulator?)
    207 208 RXA00630 GR00166 2 160 (U67196) DNA-binding response regulator [Thermotoga maritima]
    209 210 F RXA00638 GR00167 2862 3245 DNA-binding response regulator
    211 212 RXA00894 GR00244 1926 799 GTPASE-ACTIVATING PROTEIN 1
    213 214 RXA01450 GR00419 1237 1800 GTP-BINDING PROTEIN
    215 216 RXA01451 GR00419 1760 2326 GTP-BINDING PROTEIN
    217 218 RXA02376 GR00689 3064 1562 GTP-BINDING PROTEIN
    219 220 RXA01065 GR00298 2 583 GTP-BINDING PROTEIN ERA
    221 222 RXA02232 GR00653 5286 6812 GTP-BINDING PROTEIN HFLX
    223 224 RXA00848 GR00230 2125 1955 GTP-BINDING PROTEIN LEPA
    225 226 F RXA00839 GR00228 372 4 GTP-BINDING PROTEIN LEPA
    227 228 F RXA00845 GR00229 907 5 GTP-BINDING PROTEIN LEPA
    229 230 RXA02365 GR00686 1568 1029 GTP-BINDING PROTEIN LEPA
    231 232 F RXA02392 GR00696 1264 5 GTP-BINDING PROTEIN LEPA
    233 234 RXA01573 GR00438 5744 3663 2′,3′-cyclic-nucleotide 2′-posphodiesterase
    235 236 RXN01445 VV0089 14702 15694 Hypothetical Sensor Histidine Kinase (EC 2.7.3.—)
    237 238 RXN03143 VV0139 1692 2822 Hypothetical Sensor Histidine Kinase (EC 2.7.3.—)
    239 240 RXN03071 VV0040 6 344 Hypothetical Sensor Protein
    241 242 RXN03072 VV0040 396 830 Hypothetical Sensor Protein
    243 244 RXN01773 VV0015 1128 1604 PROTEIN-TYROSINE PHOSPHATASE (EC 3.1.3.48)
    245 246 RXN03090 VV0054 5296 4076 SENSORY COMPONENT OF SENSORY TRANSDUCTION HISTIDINE KINASE (EC 2.7.3.—)
    247 248 RXN00617 VV0054 4053 3826 SENSORY COMPONENT OF SENSORY TRANSDUCTION HISTIDINE KINASE (EC 2.7.3.—)
    249 250 RXN02990 VV0073 1352 1948 REGULATORY PROTEIN RECX
    251 252 RXN03100 VV0064 11866 11549 ALIPHATIC AMIDASE EXPRESSION-REGULATING PROTEIN
    253 254 RXN00031 VV0127 54780 55181 PHOSPHOHISTIDINE PHOSPHATASE SIXA (EC 3.1.3.—)
    255 256 RXN02758 VV0084 29359 28061 PHOSPHOSERINE PHOSPHATASE (EC 3.1.3.3)
    257 258 RXN00978 VV0149 1360 1974 NNRR
    259 260 RXN01349 VV0123 1531 755 REGULATORY PROTEIN BETI
    261 262 RXN00467 VV0086 60275 60943 IRON REPRESSOR
    263 264 RXN02954 VV0015 2693 3430 Hypothetical Transcriptional Regulator
    265 266 RXN03023 VV0003 6100 5744 Hypothetical Transcriptional Regulator
    267 268 RXN03127 VV0119 8276 7557 Hypothetical Transcriptional Regulator
    269 270 RXN03155 VV0186 2 1669 Hypothetical Transcriptional Regulator
    271 272 RXN01315 VV0082 13796 13146 Hypothetical Transcription Regulator
    273 274 RXN00035 VV0020 24855 24499 Hypothetical Transcriptional Regulator
    275 276 RXN00049 VV0174 11833 11147 Hypothetical Transcriptional Regulator
    277 278 RXN00486 VV0086 22816 23724 Hypothetical Transcriptional Regulator
    279 280 RXN01081 VV0084 33995 34744 Hypothetical Transcriptional Regulator
    281 282 RXN01160 VV0151 4187 3213 Hypothetical Transcriptional Regulator
    283 284 RXN02097 VV0298 184 3555 Hypothetical Transcriptional Regulator
    285 286 RXN02266 VV0020 9528 10040 Hypothetical Transcriptional Regulator
    287 288 RXN02362 VV0051 11237 7539 Hypothetical Transcriptional Regulator
    289 290 RXN02506 VV0007 25030 24149 Hypothetical Transcriptional Regulator
    291 292 RXN02620 VV0129 34206 33541 Hypothetical Transcriptional Regulator
    293 294 RXN00826 VV0180 2580 3110 Hypothetical Transcriptional Regulator
    295 296 RXS00070 VV0019 32468 32899 FERRIC UPTAKE REGULATION PROTEIN
    297 298 RXS00133 VV0046 201 1013 NITRATE/NITRITE RESPONSE REGULATOR PROTEIN NARP
    299 300 RXS00144 VV0134 20478 21053 PYRIMIDINE OPERON REGULATORY PROTEIN PYRR
    301 302 RXS00205 VV0096 4885 3779 CCPA PROTEIN
    303 304 RXS00470 VV0086 27401 28669 NITRATE/NITRITE SENSOR PROTEIN NARX (EC 2.7.3.—)
    305 306 RXS00471 VV0086 28715 29404 NITRATE/NITRITE RESPONSE REGULATOR PROTEIN NARL
    307 308 RXS00481 VV0086 43354 43938 Hypothetical Protein
    309 310 RXS00649 VV0109 10679 10224 Hypothetical Cytosolic Protein
    311 312 RXS00650 VV0109 9485 10120 NITRATE/NITRITE RESPONSE REGULATOR PROTEIN NARP
    313 314 RXS00657 VV0109 2620 3522 ACR Protein
    315 316 RXS00719 VV0232 7281 5653 Hypothetical GTP-Binding Protein
    317 318 RXS00738 VV0254 3 365 Hypothetical Cytosolic Protein
    319 320 RXS01082 VV0084 35406 34747 IRON REPRESSOR
    321 322 RXS01123 VV0143 24824 25270 Hypothetical Protein
    323 324 RXS01189 VV0169 6366 6974 NITRATE/NITRITE RESPONSE REGULATOR PROTEIN NARP
    325 326 RXS01242 VV0068 17647 16871 GLYCEROL-3-PHOSPHATE REGULON REPRESSOR
    327 328 RXS01607 VV0139 2822 3451 NITRATE/NITRITE RESPONSE REGULATOR PROTEIN NARP
    329 330 RXS01674 PROBABLE HYDROGEN PEROXIDE-INDUCIBLE GENES ACTIVATOR
    331 332 RXS01872 VV0248 2141 2968 TRANSCRIPTIONAL REGULATORY PROTEIN
    333 334 RXS02117 VV0102 8076 8549 Hypothetical Cytosolic Protein
    335 336 RXS02288 VV0127 51473 50628 GLYCEROL-3-PHOSPHATE REGULON REPRESSOR
    337 338 RXS02573 VV0098 2475 2918 ACR Protein
    339 340 RXS02627 VV0314 2981 2139 DTXR/IRON-REGULATED LIPOPROTEIN PRECURSOR
    341 342 RXS02691 VV0098 55962 56768 FATTY ACYL RESPONSIVE REGULATOR
    343 344 RXS02730 VV0145 7640 8677 RIBOSE OPERON REPRESSOR
    345 346 RXS02818 VV0347 611 6 Hypothetical Protein
    347 348 RXS02911 VV0135 24643 25101 Hypothetical Cytosolic Protein
    349 350 RXS03066 VV0038 7298 6636 Hypothetical Protein
    351 352 RXS03208 DIPHTHERIA TOXIN REPRESSOR
    353 354 F RXA00307 GR00052 467 6 DIPHTHERIA TOXIN REPRESSOR
    355 356 RXS03219 LACI-FAMILY TRANSCRIPTION REGULATOR
    357 358 F RXA02763 GR00768 1603 2586 MALTOSE OPERON TRANSCRIPTIONAL REPRESSOR
    359 360 RXS03200 PROBABLE HYDROGEN PEROXIDE-INDUCIBLE GENES ACTIVATOR
  • TABLE 2
    GENES IDENTIFIED FROM GENBANK
    GenBank ™
    Accession No. Gene Name Gene Function Reference
    A09073 ppg Phosphoenol pyruvate carboxylase Bachmann, B. et al. “DNA fragment coding for phosphoenolpyruvat
    corboxylase, recombinant DNA carrying said fragment, strains carrying the
    recombinant DNA and method for producing L-amino acids using said
    strains,” Patent: EP 0358940-A 3 Mar. 21, 1990
    A45579, Threonine dehydratase Moeckel, B. et al. “Production of L-isoleucine by means of recombinant
    A45581, micro-organisms with deregulated threonine dehydratase,” Patent: WO
    A45583, 9519442-A 5 Jul. 20, 1995
    A45585
    A45587
    AB003132 murC; ftsQ; ftsZ Kobayashi, M. et al. “Cloning, sequencing, and characterization of the ftsZ
    gene from coryneform bacteria,” Biochem. Biophys. Res. Commun.,
    236(2): 383-388 (1997)
    AB015023 murC; ftsQ Wachi, M. et al. “A murC gene from Coryneform bacteria,” Appl. Microbiol.
    Biotechnol., 51(2): 223-228 (1999)
    AB018530 dtsR Kimura, E. et al. “Molecular cloning of a novel gene, dtsR, which rescues the
    detergent sensitivity of a mutant derived from Brevibacterium lactofermentum,” Biosci. Biotechnol. Biochem.,
    60(10): 1565-1570 (1996)
    AB018531 dtsR1; dtsR2
    AB020624 murI D-glutamate racemase
    AB023377 tkt transketolase
    AB024708 gltB; gltD Glutamine 2-oxoglutarate aminotransferase large and small
    subunits
    AB025424 acn aconitase
    AB027714 rep Replication protein
    AB027715 rep; aad Replication protein; aminoglycoside adenyltransferase
    AF005242 argC N-acetylglutamate-5-semialdehyde
    dehydrogenase
    AF005635 glnA Glutamine synthetase
    AF030405 hisF cyclase
    AF030520 argG Argininosuccinate synthetase
    AF031518 argF Ornithine carbamolytransferase
    AF036932 aroD 3-dehydroquinate dehydratase
    AF038548 pyc Pyruvate carboxylase
    AF038651 dciAE; apt; rel Dipeptide-binding protein; adenine Wehmeier, L. et al. “The role of the Corynebacterium glutamicum rel gene in
    phosphoribosyltransferase; GTP pyrophosphokinase (p)ppGpp metabolism,” Microbiology, 144: 1853-1862 (1998)
    AF041436 argR Arginine repressor
    AF045998 impA Inositol monophosphate phosphatase
    AF048764 argH Argininosuccinate lyase
    AF049897 argC; argJ; argB; N-acetylglutamylphosphate reductase;
    argD; argF; argR; ornithine acetyltransferase; N-
    argG; argH acetylglutamate kinase; acetylornithine
    transminase; ornithine carbamoyltransferase; arginine repressor;
    argininosuccinate synthase; argininosuccinate lyase
    AF050109 inhA Enoyl-acyl carrier protein reductase
    AF050166 hisG ATP phosphoribosyltransferase
    AF051846 hisA Phosphoribosylformimino-5-amino-1-
    phosphoribosyl-4-imidazolecarboxamide
    isomerase
    AF052652 metA Homoserine O-acetyltransferase Park, S. et al. “Isolation and analysis of metA, a methionine biosynthetic gene
    encoding homoserine acetyltransferase in Corynebacterium glutamicum,” Mol. Cells., 8(3):
    286-294 (1998)
    AF053071 aroB Dehydroquinate synthetase
    AF060558 hisH Glutamine amidotransferase
    AF086704 hisE Phosphoribosyl-ATP-
    pyrophosphohydrolase
    AF114233 aroA 5-enolpyruvylshikimate 3-phosphate
    synthase
    AP116184 panD L-aspartate-alpha-decarboxylase precursor Dusch, N. et al. “Expression of the Corynebacterium glutamicum panD gene
    encoding L-aspartate-alpha-decarboxylase leads to pantothenate
    overproduction in Escherichia coli,” Appl. Environ. Microbiol., 65(4)1530-1539 (1999)
    AF124518 aroD; aroE 3-dehydroquinase; shikimate
    dehydrogenase
    AF124600 aroC; aroK; aroB; Chorismate synthase; shikimate kinase; 3-
    pepQ dehydroquinate synthase; putative
    cytoplasmic peptidase
    AF145897 inhA
    AF145898 inhA
    AJ001436 ectP Transport of ectoine, glycine betaine, Peter, H. et al. “Corynebacterium glutamicum is equipped with four secondary
    proline carriers for compatible solutes: Identification, sequencing, and characterization
    of the proline/ectoine uptake system, ProP, and the ectoine/proline/glycine betaine carrier,
    EctP,” J. Bacteriol., 180(22): 6005-6012 (1998)
    AJ004934 dapD Tetrahydrodipicolinate succinylase Wehrmann, A. et al. “Different modes of diaminopimelate synthesis and their
    (incompletei) role in cell wall integrity: A study with Corynebacterium glutamicum,” J. Bacteriol., 180(12):
    3159-3165 (1998)
    AJ007732 ppc; secG; amt; ocd; Phosphoenolpyruvate-carboxylase; ?; high
    soxA affinity ammonium uptake protein; putative
    ornithine-cyclodecarboxylase; sarcosine
    oxidase
    AJ010319 ftsY, glnB, glnD; srp; Involved in cell division; PII protein; Jakoby, M. et al. “Nitrogen regulation in Corynebacterium glutamicum;
    amtP uridylyltransferase (uridylyl-removing Isolation of genes involved in biochemical characterization of corresponding
    enzmye); signal recognition particle; low proteins,” FEMS Microbiol., 173(2): 303-310 (1999)
    affinity ammonium uptake protein
    AJ132968 cat Chloramphenicol aceteyl transferase
    AJ224946 mqo L-malate: quinone oxidoreductase Molenaar, D. et al. “Biochemical and genetic characterization of the
    membrane-associated malate dehydrogenase (acceptor) from Corynebacterium
    glutamicum,” Eur. J. Biochem., 254(2): 395-403 (1998)
    AJ238250 ndh NADH dehydrogenase
    AJ238703 porA Porin Lichtinger, T. et al. “Biochemical and biophysical characterization of the cell
    wall porin of Corynebacterium glutamicum: The channel is formed by a low
    molecular mass polypeptide,” Biochemistry, 37(43): 15024-15032 (1998)
    D17429 Transposable element IS31831 Vertes, A. A. et al. “Isolation and characterization of IS31831, a transposable
    element from Corynebacterium glutamicum,” Mol. Microbiol., 11(4): 739-746
    (1994)
    D84102 odhA 2-oxoglutarate dehydrogenase Usuda, Y. et al. “Molecular cloning of the Corynebacterium glutamicum
    (Brevibacterium lactofermentum AJ12036) odhA gene encoding a novel type
    of 2-oxoglutarate dehydrogenase,” Microbiology, 142: 3347-3354 (1996)
    E01358 hdh; hk Homoserine dehydrogenase; homoserine Katsumata, R. et al. “Production of L-thereonine and L-isoleucine,” Patent: JP
    kinase 1987232392-A 1 Oct. 12, 1987
    E01359 Upstream of the start codon of homoserine Katsumata, R. et al. “Production of L-thereonine and L-isoleucine,” Patent: JP
    kinase gene 1987232392-A 2 Oct. 12, 1987
    E01375 Tryptophan operon
    E01376 trpL; trpE Leader peptide; anthranilate synthase Matsui, K. et al. “Tryptophan operon, peptide and protein coded thereby,
    utilization of tryptophan operon gene expression and production of
    tryptophan,” Patent: JP 1987244382-A 1 Oct. 24, 1987
    E01377 Promoter and operator regions of Matsui, K. et al. “Tryptophan operon, peptide and protein coded thereby,
    tryptophan operon utilization of tryptophan operon gene expression and production of
    tryptophan,” Patent: JP 1987244382-A 1 Oct. 24, 1987
    E03937 Biotin-synthase Hatakeyama, K. et al. “DNA fragment containing gene capable of coding
    biotin synthetase and its utilization,” Patent: JP 1992278088-A 1 Oct. 02, 1992
    E04040 Diamino pelargonic acid aminotransferase Kohama, K. et al. “Gene coding diaminopelargonic acid aminotransferase and
    desthiobiotin synthetase and its utilization,” Patent: JP 1992330284-A 1
    Nov. 18, 1992
    E04041 Desthiobiotinsynthetase Kohama, K. et al. “Gene coding diaminopelargonic acid aminotransferase and
    desthiobiotin synthetase and its utilization,” Patent: JP 1992330284-A 1
    Nov. 18, 1992
    E04307 Flavum aspartase Kurusu, Y. et al. “Gene DNA coding aspartase and utilization thereof,” Patent:
    JP 1993030977-A 1 Feb. 09, 1993
    E04376 Isocitric acid lyase Katsumata, R. et al. “Gene manifestation controlling DNA,” Patent: JP
    1993056782-A 3 Mar. 09, 1993
    E04377 Isocitric acid lyase N-terminal fragment Katsumata, R. et al. “Gene manifestation controlling DNA,” Patent: JP
    1993056782-A 3 Mar. 09, 1993
    E04484 Prephenate dehydratase Sotouchi, N. et al. “Production of L-phenylalanine by fermentation,” Patent: JP
    1993076352-A 2 Mar. 30, 1993
    E05108 Aspartokinase Fugono, N. et al. “Gene DNA coding Aspartokinase and its use,” Patent: JP
    1993184366-A 1 Jul. 27, 1993
    E05112 Dihydro-dipichorinate synthetase Hatakeyama, K. et al. “Gene DNA coding dihydrodipicolinic acid synthetase
    and its use,” Patent: JP 1993184371-A 1 Jul. 27, 1993
    E05776 Diaminopimelic acid dehydrogenase Kobayashi, M. et al. “Gene DNA coding Diaminopimelic acid dehydrogenase
    and its use,” Patent: JP 1993284970-A 1 Nov. 02, 1993
    E05779 Threonine synthase Kohama, K. et al. “Gene DNA coding threonine synthase and its use,” Patent:
    JP 1993284972-A 1 Nov. 02, 1993
    E06110 Prephenate dehydratase Kikuchi, T. et al. “Production of L-phenylalanine by fermentation method,”
    Patent: JP 1993344881-A 1 Dec. 27, 1993
    E06111 Mutated Prephenate dehydratase Kikuchi, T. et al. “Production of L-phenylalanine by fermentation method,”
    Patent: JP 1993344881-A 1 Dec. 27, 1993
    E06146 Acetohydroxy acid synthetase Inui, M. et al. “Gene capable of coding Acetohydroxy acid synthetase and its
    use,” Patent: JP 1993344893-A 1 Dec. 27, 1993
    E06825 Aspartokinase Sugimoto, M. et al. “Mutant aspartokinase gene,” patent: JP 1994062866-A 1
    Mar. 08, 1994
    E06826 Mutated aspartokinase alpha subunit Sugimoto, M. et al. “Mutant aspartokinase gene,” patent: JP 1994062866-A 1
    Mar. 08, 1994
    E06827 Mutated aspartokinase alpha subunit Sugimoto, M. et al. “Mutant aspartokinase gene,” patent: JP 1994062866-A 1
    Mar. 08, 1994
    E07701 secY Honno, N. et al. “Gene DNA participating in integration of membraneous
    protein to membrane,” Patent: JP 1994169780-A 1 Jun. 21, 1994
    E08177 Aspartokinase Sato, Y. et al. “Genetic DNA capable of coding Aspartokinase released from
    feedback inhibition and its utilization,” Patent: JP 1994261766-A 1 Sep. 20, 1994
    E08178, Feedback inhibition-released Aspartokinase Sato, Y. et al. “Genetic DNA capable of coding Aspartokinase released from
    E08179, feedback inhibition and its utilization,” Patent: JP 1994261766-A 1 Sep. 20, 1994
    E08180,
    E08181,
    E08182
    E08232 Acetohydroxy-acid isomeroreductase Inui, M. et al. “Gene DNA coding acetohydroxy acid isomeroreductase,”
    Patent: JP 1994277067-A 1 Oct. 04, 1994
    E08234 secE Asai, Y. et al. “Gene DNA coding for translocation machinery of protein,”
    Patent: JP 1994277073-A 1 Oct. 04, 1994
    E08643 FT aminotransferase and desthiobiotin Hatakeyama, K. et al. “DNA fragment having promoter function in
    synthetase promoter region coryneform bacterium,” Patent: JP 1995031476-A 1 Feb. 03, 1995
    E08646 Biotin synthetase Hatakeyama, K. et al. “DNA fragment having promoter function in
    coryneform bacterium,” Patent: JP 1995031476-A 1 Feb. 03, 1995
    E08649 Aspartase Kohama, K. et al “DNA fragment having promoter function in coryneform
    bacterium,” Patent: JP 1995031478-A 1 Feb. 03, 1995
    E08900 Dihydrodipicolinate reductase Madori, M. et al. “DNA fragment containing gene coding Dihydrodipicolinate
    acid reductase and utilization thereof,” Patent: JP 1995075578-A 1 Mar. 20, 1995
    E08901 Diaminopimelic acid decarboxylase Madori, M. et al. “DNA fragment containing gene coding Diaminopimelic acid
    decarboxylase and utilization thereof,” Patent: JP 1995075579-A 1 Mar. 20, 1995
    E12594 Serine hydroxymethyltransferase Hatakeyama, K. et al. “Production of L-trypophan,” Patent: JP 1997028391-A
    1 Feb. 04, 1997
    E12760, transposase Moriya, M. et al. “Amplification of gene using artificial transposon,” Patent:
    E12759, JP 1997070291-A Mar. 18, 1997
    E12758
    E12764 Arginyl-tRNA synthetase; diaminopimelic Moriya, M. et al. “Amplification of gene using artificial transposon,” Patent:
    acid decarboxylase JP 1997070291-A Mar. 18, 1997
    E12767 Dihydrodipicolinic acid synthetase Moriya, M. et al. “Amplification of gene using artificial transposon,” Patent:
    JP 1997070291-A Mar. 18, 1997
    E12770 aspartokinase Moriya, M. et al. “Amplification of gene using artificial transposon,” Patent:
    JP 1997070291-A Mar. 18, 1997
    E12773 Dihydrodipicolinic acid reductase Moriya, M. et al. “Amplification of gene using artificial transposon,” Patent:
    JP 1997070291-A Mar. 18, 1997
    E13655 Glucose-6-phosphate dehydrogenase Hatakeyama, K. et al. “Glucose-6-phosphate dehydrogenase and DNA capable
    of coding the same,” Patent: JP 1997224661-A 1 Sep. 02, 1997
    L01508 IlvA Threonine dehydratase Moeckel, B. et al. “Functional and structural analysis of the threonine
    dehydratase of Corynebacterium glutamicum,” J. Bacteriol., 174: 8065-8072
    (1992)
    L07603 EC 4.2.1.15 3-deoxy-D-arabinoheptulosonate-7- Chen, C. et al. “The cloning and nucleotide sequence of Corynebacterium
    phosphate synthase glutamicum 3-deoxy-D-arabinoheptulosonate-7-phosphate synthase gene,”
    FEMS Microbiol. Lett., 107: 223-230 (1993)
    L09232 IlvB; ilvN; ilvC Acetohydroxy acid synthase large subunit; Keilhauer, C. et al. “Isoleucine synthesis in Corynebacterium glutamicum:
    Acetohydroxy acid synthase small subunit; molecular analysis of the ilvB-ilvN-ilvC operon,” J. Bacteriol., 175(17): 5595-5603
    Acetohydroxy acid isomeroreductase (1993)
    L18874 PtsM Phosphoenolpyruvate sugar Fouet, A et al. “Bacillus subtilis sucrose-specific enzyme II of the
    phosphotransferase phosphotransferase system: expression in Escherichia coli and homology to
    enzymes II from enteric bacteria,” PNAS USA, 84(24): 8773-8777 (1987); Lee, J. K.
    et al. “Nucleotide sequence of the gene encoding the Corynebacterium
    glutamicum mannose enzyme II and analyses of the deduced protein
    sequence,” FEMS Microbiol. Lett., 119(1-2): 137-145 (1994)
    L27123 aceB Malate synthase Lee, H-S. et al. “Molecular characterization of aceB, a gene encoding malate
    synthase in Corynebacterium glutamicum,” J. Microbiol. Biotechnol.,
    4(4): 256-263 (1994)
    L27126 Pyruvate kinase Jetten, M. S. et al. “Structural and functional analysis of pyruvate kinase from
    Corynebacterium glutamicum,” Appl. Environ. Microbiol., 60(7): 2501-2507
    (1994)
    L28760 aceA Isocitrate lyase
    L35906 dtxr Diphtheria toxin repressor Oguiza, J. A. et al. “Molecular cloning, DNA sequence analysis, and
    characterization of the Corynebacterium diphtheriae dtxR from Brevibacterium
    lactofermentum,” J. Bacteriol., 177(2): 465-467 (1995)
    M13774 Prephenate dehydratase Follettie, M. T. et al. “Molecular cloning and nucleotide sequence of the
    Corynebacterium glutamicum pheA gene,” J. Bacteriol., 167: 695-702 (1986)
    M16175 5S rRNA Park, Y-H. et al. “Phylogenetic analysis of the coryneform bacteria by 56
    rRNA sequences,” J. Bacteriol., 169: 1801-1806 (1987)
    M16663 trpE Anthranilate synthase, 5′ end Sano, K. et al. “Structure and function of the trp operon control regions of
    Brevibacterium lactofermentum, a glutamic-acid-producing bacterium,” Gene,
    52: 191-200 (1987)
    M16664 trpA Tryptophan synthase, 3′ end Sano, K. et al. “Structure and function of the trp operon control regions of
    Brevibacterium lactofermentum, a glutamic-acid-producing bacterium,” Gene,
    52: 191-200 (1987)
    M25819 Phosphoenolpyruvate carboxylase O'Regan, M. et al. “Cloning and nucleotide sequence of the
    Phosphoenolpyruvate carboxylase-coding gene of Corynebacterium
    glutamicum ATCC13032,” Gene, 77(2): 237-251 (1989)
    M85106 23S rRNA gene insertion sequence Roller, C. et al. “Gram-positive bacteria with a high DNA G + C content are
    characterized by a common insertion within their 23S rRNA genes,” J. Gen.
    Microbiol., 138: 1167-1175 (1992)
    M85107, 23S rRNA gene insertion sequence Roller, C. et al. “Gram-positive bacteria with a high DNA G + C content are
    M85108 characterized by a common insertion within their 23S rRNA genes,” J. Gen.
    Microbiol., 138: 1167-1175 (1992)
    M89931 aecD; brnQ; yhbw Beta C-S lyase; branched-chain amino acid Rossol, I. et al. “The Corynebacterium glutamicum aecD gene encodes a C-S
    uptake carrier; hypothetical protein yhbw lyase with alpha, beta-elimination activity that degrades aminoethylcysteine,”
    J. Bacteriol., 174(9): 2968-2977 (1992); Tauch, A. et al. “Isoleucine uptake in
    Corynebacterium glutamicum ATCC 13032 is directed by the brnQ gene
    product,” Arch. Microbiol., 169(4): 303-312 (1998)
    S59299 trp Leader gene (promoter) Herry, D. M. et al. “Cloning of the trp gene cluster from a tryptophan-
    hyperproducing strain of Corynebacterium glutamicum: identification of a
    mutation in the trp leader sequence,” Appl. Environ. Microbiol., 59(3): 791-799
    (1993)
    U11545 trpD Anthranilate phosphoribosyltransferase O'Gara, J. P. and Dunican, L. K. (1994) Complete nucleotide sequence of the
    Corynebacterium glutamicum ATCC 21850 tpD gene.” Thesis, Microbiology
    Department, University College Galway, Ireland.
    U13922 cglIM; cglIR; clgIIR Putative type II 5-cytosoine Schafer, A. et al. “Cloning and characterization of a DNA region encoding a
    methyltransferase; putative type II stress-sensitive restriction system from Corynebacterium glutamicum ATCC
    restriction endonuclease; putative type I or 13032 and analysis of its role in intergeneric conjugation with Escherichia
    type III restriction endonuclease coli,” J. Bacteriol., 176(23): 7309-7319 (1994); Schafer, A. et al. “The
    Corynebacterium glutamicum cglIM gene encoding a 5-cytosine in an McrBC-
    deficient Escherichia coli strain,” Gene, 203(2): 95-101 (1997)
    U14965 recA
    U31224 ppx Ankri, S. et al. “Mutations in the Corynebacterium glutamicumproline
    biosynthetic pathway: A natural bypass of the proA step,” J. Bacteriol.,
    178(15): 4412-4419 (1996)
    U31225 proC L-proline: NADP+ 5-oxidoreductase Ankri, S. et al. “Mutations in the Corynebacterium glutamicumproline
    biosynthetic pathway: A natural bypass of the proA step,” J. Bacteriol.,
    178(15): 4412-4419 (1996)
    U31230 obg; proB; unkdh ?; gamma glutamyl kinase; similar to D- Ankri, S. et al. “Mutations in the Corynebacterium glutamicumproline
    isomer specific 2-hydroxyacid biosynthetic pathway: A natural bypass of the proA step,” J. Bacteriol.,
    dehydrogenases 178(15): 4412-4419 (1996)
    U31281 bioB Biotin synthase Serebriiskii, I. G., “Two new members of the bio B superfamily: Cloning,
    sequencing and expression of bio B genes of Methylobacillus flagellatum and
    Corynebacterium glutamicum,” Gene, 175: 15-22 (1996)
    U35023 thtR; accBC Thiosulfate sulfurtransferase; acyl CoA Jager, W. et al. “A Corynebacterium glutamicum gene encoding a two-domain
    carboxylase protein similar to biotin carboxylases and biotin-carboxyl-carrier proteins,”
    Arch. Microbiol., 166(2); 76-82 (1996)
    U43535 cmr Multidrug resistance protein Jager, W. et al. “A Corynebacterium glutamicum gene conferring multidrug
    resistance in the heterologous host Escherichia coli,” J. Bacteriol.,
    179(7): 2449-2451 (1997)
    U43536 clpB Heat shock ATP-binding protein
    U53587 aphA-3 3′5″-aminoglycoside phosphotransferase
    U89648 Corynebacterium glutamicum unidentified
    sequence involved in histidine biosynthesis,
    partial sequence
    X04960 trpA; trpB; trpC; trpD; Tryptophan operon Matsui, K. et al. “Complete nucleotide and deduced amino acid sequences of
    trpE; trpG; trpL the Brevibacterium lactofermentum tryptophan operon,” Nucleic Acids Res.,
    14(24): 10113-10114 (1986)
    X07563 lys A DAP decarboxylase (meso-diaminopimelate Yeh, P. et al. “Nucleic sequence of the lysA gene of Corynebacterium
    decarboxylase, EC 4.1.1.20) glutamicum and possible mechanisms for modulation of its expression,” Mol.
    Gen. Genet., 212(1): 112-119 (1988)
    X14234 EC 4.1.1.31 Phosphoenolpyruvate carboxylase Eikmanns, B. J. et al. “The Phosphoenolpyruvate carboxylase gene of
    Corynebacterium glutamicum: Molecular cloning, nucleotide sequence, and
    expression,” Mol. Gen. Genet., 218(2): 330-339 (1989); Lepiniec, L. et al.
    “Sorghum Phosphoenolpyruvate carboxylase gene family: structure, function
    and molecular evolution,” Plant. Mol. Biol., 21 (3): 487-502 (1993)
    X17313 fda Fructose-bisphosphate aldolase Von der Osten, C. H. et al. “Molecular cloning, nucleotide sequence and fine-
    structural analysis of the Corynebacterium glutamicum fda gene: structural
    comparison of C. glutamicum fructose-1,6-biphosphate aldolase to class I and
    class II aldolases,” Mol. Microbiol.,
    X53993 dapA L-2,3-dihydrodipicolinate synthetase (EC Bonnassie, S. et al. “Nucleic sequence of the dapA gene from
    4.2.1.52) Corynebacterium glutamicum,” Nucleic Acids Res., 18(21): 6421 (1990)
    X54223 AttB-related site Cianciotto, N. et al. “DNA sequence homology between att B-related sites of
    Corynebacterium diphtheriae, Corynebacterium ulcerans, Corynebacterium
    glutamicum, and the attP site of lambdacorynephage,” FEMS. Microbiol,
    Lett., 66: 299-302 (1990)
    X54740 argS; lysA Arginyl-tRNA synthetase; Diaminopimelate Marcel, T. et al. “Nucleotide sequence and organization of the upstream region
    decarboxylase of the Corynebacterium glutamicum lysA gene,” Mol. Microbiol., 4(11): 1819-1830
    (1990)
    X55994 trpL; trpE Putative leader peptide; anthranilate Heery, D. M. et al. “Nucleotide sequence of the Corynebacterium glutamicum
    synthase component 1 trpE gene,” Nucleic Acids Res., 18(23): 7138 (1990)
    X56037 thrC Threonine synthase Han, K. S. et al. “The molecular structure of the Corynebacterium glutamicum
    threonine synthase gene,” Mol. Microbiol., 4(10): 1693-1702 (1990)
    X56075 attB-related site Attachment site Cianciotto, N. et al. “DNA sequence homology between att B-related sites of
    Corynebacterium diphtheriae, Corynebacterium ulcerans, Corynebacterium
    glutamicum, and the attP site of lambdacorynephage,” FEMS. Microbiol,
    Lett., 66: 299-302 (1990)
    X57226 lysC-alpha; lysC-beta; Aspartokinase-alpha subunit; Kalinowski, J. et al. “Genetic and biochemical analysis of the Aspartokinase
    asd Aspartokinase-beta subunit; aspartate beta from Corynebacterium glutamicum,” Mol. Microbiol., 5(5): 1197-1204 (1991);
    semialdehyde dehydrogenase Kalinowski, J. et al. “Aspartokinase genes lysC alpha and lysC beta overlap
    and are adjacent to the aspertate beta-semialdehyde dehydrogenase gene asd in
    Corynebacterium glutamicum,” Mol. Gen. Genet., 224(3): 317-324 (1990)
    X59403 gap; pgk; tpi Glyceraldehyde-3-phosphate; Eikmanns, B. J. “Identification, sequence analysis, and expression of a
    phosphoglycerate kinase; triosephosphate Corynebacterium glutamicum gene cluster encoding the three glycolytic
    isomerase enzymes glyceraldehyde-3-phosphate dehydrogenase, 3-phosphoglycerate
    kinase, and triosephosphate isomeras,” J. Bacteriol., 174(19): 6076-6086
    (1992)
    X59404 gdh Glutamate dehydrogenase Bormann, E. R. et al. “Molecular analysis of the Corynebacterium glutamicum
    gdh gene encoding glutamate dehydrogenase,” Mol. Microbiol., 6(3): 317-326
    (1992)
    X60312 lysI L-lysine permease Seep-Feldhaus, A. H. et al. “Molecular analysis of the Corynebacterium
    glutamicum lysl gene involved in lysine uptake,” Mol. Microbiol., 5(12): 2995-3005
    (1991)
    X66078 cop1 Ps1 protein Joliff, G. et al. “Cloning and nucleotide sequence of the csp1 gene encoding
    PS1, one of the two major secreted proteins of Corynebacterium glutamicum:
    The deduced N-terminal region of PS1 is similar to the Mycobacterium antigen
    85 complex,” Mol. Microbiol., 6(16): 2349-2362 (1992)
    X66112 glt Citrate synthase Eikmanns, B. J. et al. “Cloning sequence, expression and transcriptional
    analysis of the Corynebacterium glutamicum gltA gene encoding citrate
    synthase,” Microbiol., 140: 1817-1828 (1994)
    X67737 dapB Dihydrodipicolinate reductase
    X69103 csp2 Surface layer protein PS2 Peyret, J. L. et al. “Characterization of the cspB gene encoding PS2, an ordered
    surface-layer protein in Corynebacterium glutamicum,” Mol. Microbiol.,
    9(1): 97-109 (1993)
    X69104 IS3 related insertion element Bonamy, C. et al. “Identification of IS1206, a Corynebacterium glutamicum
    IS3-related insertion sequence and phylogenetic analysis,” Mol. Microbiol.,
    14(3): 571-581 (1994)
    X70959 leuA Isopropylmalate synthase Patek, M. et al. “Leucine synthesis in Corynebacterium glutamicum: enzyme
    activities, structure of leuA, and effect of leuA inactivation on lysine
    synthesis,” Appl. Environ. Microbiol., 60(1): 133-140 (1994)
    X71489 icd Isocitrate dehydrogenase (NADP+) Eikmanns, B. J. et al. “Cloning sequence analysis, expression, and inactivation
    of the Corynebacterium glutamicum icd gene encoding isocitrate
    dehydrogenase and biochemical characterization of the enzyme,” J. Bacteriol.,
    177(3): 774-782 (1995)
    X72855 GDHA Glutamate dehydrogenase (NADP+)
    X75083, mtrA 5-methyltryptophan resistance Heery, D. M. et al. “A sequence from a tryptophan-hyperproducing strain of
    X70584 Corynebacterium glutamicum encoding resistance to 5-methyltryptophan,”
    Biochem. Biophys. Res. Commun., 201(3): 1255-1262 (1994)
    X75085 recA Fitzpatrick, R. et al. “Construction and characterization of recA mutant strains
    of Corynebacterium glutamicum and Brevibacterium lactofermentum,” Appl.
    Microbiol. Biotechnol., 42(4): 575-580 (1994)
    X75504 aceA; thiX Partial Isocitrate lyase; ? Reinscheid, D. J. et al. “Characterization of the isocitrate lyase gene from
    Corynebacterium glutamicum and biochemical analysis of the enzyme,” J.
    Bacteriol., 176(12): 3474-3483 (1994)
    X76875 ATPase beta-subunit Ludwig, W. et al. “Phylogenetic relationships of bacteria based on comparative
    sequence analysis of elongation factor Tu and ATP-synthase beta-subunit
    genes,” Antonie Van Leeuwenhoek, 64: 285-305 (1993)
    X77034 tuf Elongation factor Tu Ludwig, W. et al. “Phylogenetic relationships of bacteria based on comparative
    sequence analysis of elongation factor Tu and ATP-synthase beta-subunit
    genes,” Antonie Van Leeuwenhoek, 64: 285-305 (1993)
    X77384 recA Billman-Jacobe, H. “Nucleotide sequence of a recA gene from
    Corynebacterium glutamicum,” DNA Seq., 4(6): 403-404 (1994)
    X78491 aceB Malate synthase Reinscheid, D. J. et al. “Malate synthase from Corynebacterium glutamicum
    pta-ack operon encoding phosphotransacetylase: sequence analysis,”
    Microbiology, 140: 3099-3108 (1994)
    X80629 16S rDNA 16S ribosomal RNA Rainey, F. A. et al. “Phylogenetic analysis of the genera Rhodococcus and
    Norcardia and evidence for the evolutionary origin of the genus Norcardia
    from within the radiation of Rhodococcus species,” Microbiol., 141: 523-528
    (1995)
    X81191 gluA; gluB; gluC; Glutamate uptake system Kronemeyer, W. et al. “Structure of the gluABCD cluster encoding the
    gluD glutamate uptake system of Corynebacterium glutamicum,” J. Bacteriol.,
    177(5): 1152-1158 (1995)
    X81379 dapE Succinyldiaminopimelate desuccinylase Wehrmann, A. et al. “Analysis of different DNA fragments of
    Corynebacterium glutamicum complementing dapE of Escherichia coli,”
    Microbiology, 40: 3349-56 (1994)
    X82061 16S rDNA 16S ribosomal RNA Ruimy, R. et al. “Phylogeny of the genus Corynebacterium deduced from
    analyses of small-subunit ribosomal DNA sequences,” Int. J. Syst. Bacteriol.,
    45(4): 740-746 (1995)
    X82928 asd; lysC Aspartate-semialdehyde dehydrogenase; ? Serebrijski, I. et al. “Multicopy suppression by asd gene and osmotic stress-
    dependent complementation by heterologous proA in proA mutants,” J.
    Bacteriol., 177(24): 7255-7260 (1995)
    X82929 proA Gamma-glutamyl phosphate reductase Serebrijski, I. et al. “Multicopy suppression by asd gene and osmotic stress-
    dependent complementation by heterologous proA in proA mutants,” J.
    Bacteriol., 177(24): 7255-7260 (1995)
    X84257 16S rDNA 165 ribosomal RNA Pascual, C. et al. “Phylogenetic analysis of the genus Corynebacterium based
    on 16S rRNA gene sequences,” Int. J. Syst. Bacteriol., 45(4): 724-728 (1995)
    X85965 aroP; dapE Aromatic amino acid permease; ? Wehrmann, A. et al. “Functional analysis of sequences adjacent to dapE of
    Corynebacterium glutamicumproline reveals the presence of aroP, which
    encodes the aromatic amino acid transporter,” J. Bacteriol., 177(20): 5991-5993
    (1995)
    X86157 argB; argC; argD; Acetylglutamate kinase; N-acetyl-gamma- Sakanyan, V. et al. “Genes and enzymes of the acetyl cycle of arginine
    argF; argJ glutamyl-phosphate reductase; biosynthesis in Corynebacterium glutamicum: enzyme evolution in the early
    acetylornithine aminotransferase; ornithine steps of the arginine pathway,” Microbiology, 142: 99-108 (1996)
    carbamoyltransferase; glutamate N-
    acetyltransferase
    X89084 pta; ackA Phosphate acetyltransferase; acetate kinase Reinscheid, D. J. et al. “Cloning, sequence analysis, expression and inactivation
    of the Corynebacterium glutamicum pta-ack operon encoding
    phosphotransacetylase and acetate kinase,” Microbiology, 145: 503-513 (1999)
    X89850 attB Attachment site Le Marrec, C. et al. “Genetic characterization of site-specific integration
    functions of phi AAU2 infecting “Arthrobacter aureus C70,” J. Bacteriol.,
    178(7): 1996-2004 (1996)
    X90356 Promoter fragment F1 Patek, M. et al. “Promoters from Corynebacterium glutamicum: cloning,
    molecular analysis and search for a consensus motif,” Microbiology,
    142: 1297-1309 (1996)
    X90357 Promoter fragment F2 Patek, M. et al. “Promoters from Corynebacterium glutamicum: cloning,
    molecular analysis and search for a consensus motif,” Microbiology,
    142: 1297-1309 (1996)
    X90358 Promoter fragment F10 Patek, M. et al. “Promoters from Corynebacterium glutamicum: cloning,
    molecular analysis and search for a consensus motif,” Microbiology,
    142: 1297-1309 (1996)
    X90359 Promoter fragment F13 Patek, M. et al. “Promoters from Corynebacterium glutamicum: cloning,
    molecular analysis and search for a consensus motif,” Microbiology,
    142: 1297-1309 (1996)
    X90360 Promoter fragment F22 Patek, M. et al. “Promoters from Corynebacterium glutamicum: cloning,
    molecular analysis and search for a consensus motif,” Microbiology,
    142: 1297-1309 (1996)
    X90361 Promoter fragment F34 Patek, M. et al. “Promoters from Corynebacterium glutamicum: cloning,
    molecular analysis and search for a consensus motif,” Microbiology,
    142: 1297-1309 (1996)
    X90362 Promoter fragment F37 Patek, M. et al. “Promoters from Corynebacterium glutamicum: cloning,
    molecular analysis and search for a consensus motif,” Microbiology,
    142: 1297-1309 (1996)
    X90363 Promoter fragment F45 Patek, M. et al. “Promoters from Corynebacterium glutamicum: cloning,
    molecular analysis and search for a consensus motif,” Microbiology,
    142: 1297-1309 (1996)
    X90364 Promoter fragment F64 Patek, M. et al. “Promoters from Corynebacterium glutamicum: cloning,
    molecular analysis and search for a consensus motif,” Microbiology,
    142: 1297-1309 (1996)
    X90365 Promoter fragment F75 Patek, M. et al. “Promoters from Corynebacterium glutamicum: cloning,
    molecular analysis and search for a consensus motif,” Microbiology,
    142: 1297-1309 (1996)
    X90366 Promoter fragment PF101 Patek, M. et al. “Promoters from Corynebacterium glutamicum: cloning,
    molecular analysis and search for a consensus motif,” Microbiology,
    142: 1297-1309 (1996)
    X90367 Promoter fragment PF104 Patek, M. et al. “Promoters from Corynebacterium glutamicum: cloning,
    molecular analysis and search for a consensus motif,” Microbiology,
    142: 1297-1309 (1996)
    X90368 Promoter fragment PF109 Patek, M. et al. “Promoters from Corynebacterium glutamicum: cloning,
    molecular analysis and search for a consensus motif,” Microbiology,
    142: 1297-1309 (1996)
    X93513 amt Ammonium transport system Siewe, R. M. et al. “Functional and genetic characterization of the (methyl)
    ammonium uptake carrier of Corynebacterium glutamicum,” J. Biol. Chem.,
    271(10): 5398-5403 (1996)
    X93514 betP Glycine betaine transport system Peter, H. et al. “Isolation, characterization, and expression of the
    Corynebacterium glutamicum betP gene, encoding the transport system for the
    compatible solute glycine betaine,” J. Bacteriol., 178(17): 5229-5234 (1996)
    X95649 orf4 Patek, M. et al. “Identification and transcriptional analysis of the dapB-ORF2-
    dapA-ORF4 operon of Corynebacterium glutamicum, encoding two enzymes
    involved in L-lysine synthesis,” Biotechnol. Lett., 19: 1113-1117 (1997)
    X96471 lysE; lysG Lysine exporter protein; Lysine export Vrljic, M. et al. “A new type of transporter with a new type of cellular
    regulator protein function: L-lysine export from Corynebacterium glutamicum,” Mol.
    Microbiol., 22(5): 815-826 (1996)
    X96580 panB; panC; xylB 3-methyl-2-oxobutanoate Sahm, H. et al. “D-pantothenate synthesis in Corynebacterium glutamicum and
    hydroxymethyltransferase; pantoate-beta- use of panBC and genes encoding L-valine synthesis for D-pantothenate
    alanine ligase; xylulokinase overproduction,” Appl. Environ. Microbiol., 65(5): 1973-1979 (1999)
    X96962 Insertion sequence IS1207 and transposase
    X99289 Elongation factor P Ramos, A. et al. “Cloning, sequencing and expression of the gene encoding
    elongation factor P in the amino-acid producer Brevibacterium lactofermentum
    (Corynebacterium glutamicum ATCC 13869),” Gene, 198: 217-222 (1997)
    Y00140 thrB Homoserine kinase Mateos, L. M. et al. “Nucleotide sequence of the homoserine kinase (thrB) gene
    of the Brevibacterium lactofermentum,” Nucleic Acids Res., 15(9): 3922 (1987)
    Y00151 ddh Meso-diaminopimelate D-dehydrogenase Ishino, S. et al. “Nucleotide sequence of the meso-diaminopimelate D-
    (EC 1.4.1.16) dehydrogenase gene from Corynebacterium glutamicum,” Nucleic Acids Res.,
    15(9): 3917 (1987)
    Y00476 thrA Homoserine dehydrogenase Mateos, L. M. et al. “Nucleotide sequence of the homoserine dehydrogenase
    (thrA) gene of the Brevibacterium lactofermentum,” Nucleic Acids Res.,
    15(24): 10598 (1987)
    Y00546 hom; thrB Homoserine dehydrogenase; homoserine Peoples, O. P. et al. “Nucleotide sequence and fine structural analysis of the
    kinase Corynebacterium glutamicum hom-thrB operon,” Mol. Microbiol., 2(1): 63-72
    (1988)
    Y08964 murC; ftsQ/divD; ftsZ UPD-N-acetylmuramate-alanine ligase; Honrubia, M. P. et al. “Identification, characterization, and chromosomal
    division initiation protein or cell division organization of the ftsZ gene from Brevibacterium lactofermentum,” Mol. Gen.
    protein; cell division protein Genet., 259(1): 97-104 (1998)
    Y09163 putP High affinity proline transport system Peter, H. et al. “Isolation of the putP gene of Corynebacterium
    glutamicumproline and characterization of a low-affinity uptake system for
    compatible solutes,” Arch. Microbiol., 168(2): 143-151 (1997)
    Y09548 pyc Pyruvate carboxylase Peters-Wendisch, P. G. et al. “Pyruvate carboxylase from Corynebacterium
    glutamicum: characterization, expression and inactivation of the pyc gene,”
    Microbiology, 144: 915-927 (1998)
    Y09578 leuB 3-isopropylmalate dehydrogenase Patek, M. et al. “Analysis of the leuB gene from Corynebacterium
    glutamicum,” Appl. Microbiol. Biotechnol., 50(1): 42-47 (1998)
    Y12472 Attachment site bacteriophage Phi-16 Moreau, S. et al. “Site-specific integration of corynephage Phi-16: The
    construction of an integration vector,” Microbiol., 145: 539-548 (1999)
    Y12537 proP Proline/ectoine uptake system protein Peter, H. et al. “Corynebacterium glutamicum is equipped with four secondary
    carriers for compatible solutes: Identification, sequencing, and characterization
    of the proline/ectoine uptake system, ProP, and the ectoine/proline/glycine
    betaine carrier, EctP,” J. Bacteriol., 180(22): 6005-6012 (1998)
    Y13221 glnA Glutamine synthetase I Jakoby, M. et al. “Isolation of Corynebacterium glutamicum glnA gene
    encoding glutamine synthetase I,” FEMS Microbiol. Lett., 154(1): 81-88 (1997)
    Y16642 lpd Dihydrolipoamide dehydrogenase
    Y18059 Attachment site Corynephage 304L Moreau, S. et al. “Analysis of the integration functions of φ 304L: An
    integrase module among corynephages,” Virology, 255(1): 150-159 (1999)
    Z21501 argS; lysA Arginyl-tRNA synthetase; diaminopimelate Oguiza, J. A. et al. “A gene encoding arginyl-tRNA synthetase is located in the
    decarboxylase (partial) upstream region of the lysA gene in Brevibacterium lactofermentum:
    Regulation of argS-lysA cluster expression by arginine,” J.
    Bacteriol., 175(22): 7356-7362 (1993)
    Z21502 dapA; dapB Dihydrodipicolinate synthase; Pisabarro, A. et al. “A cluster of three genes (dapA, orf2, and dapB) of
    dihydrodipicolinate reductase Brevibacterium lactofermentum encodes dihydrodipicolinate reductase, and a
    third polypeptide of unknown function,” J. Bacteriol., 175(9): 2743-2749
    (1993)
    Z29563 thrC Threonine synthase Malumbres, M. et al. “Analysis and expression of the thrC gene of the encoded
    threonine synthase,” Appl. Environ. Microbiol., 60(7)2209-2219 (1994)
    Z46753 16S rDNA Gene for 16S ribosomal RNA
    Z49822 sigA SigA sigma factor Oguiza, J. A. et al “Multiple sigma factor genes in Brevibacterium
    lactofermentum: Characterization of sigA and sigB,” J. Bacteriol., 178(2): 550-553
    (1996)
    Z49823 galE; dtxR Catalytic activity UDP-galactose 4- Oguiza, J. A. et al “The galE gene encoding the UDP-galactose 4-epimerase of
    epimerase; diphtheria toxin regulatory Brevibacterium lactofermentum is coupled transcriptionally to the dmdR
    protein gene,” Gene, 177: 103-107 (1996)
    Z49824 orf1; sigB ?; SigB sigma factor Oguiza, J. A. et al “Multiple sigma factor genes in Brevibacterium
    lactofermentum: Characterization of sigA and sigB,” J. Bacteriol., 178(2): 550-553
    (1996)
    Z66534 Transposase Correia, A. et al. “Cloning and characterization of an IS-like element present in
    the genome of Brevibacterium lactofermentum ATCC 13869,” Gene,
    170(1): 91-94 (1996)

    1A sequence for this gene was published in the indicated reference. However, the sequence obtained by the inventors of the present application is significantly longer than the published version. It is believed that the published version relied on an incorrect start codon, and thus represents only a fragment of the actual coding region.
  • TABLE 3
    Corynebacterium and Brevibacterium Strains Which May be Used in the Practice of the Invention
    Genus species ATCC FERM NRRL CECT NCIMB CBS NCTC DSMZ
    Brevibacterium ammoniagenes 21054
    Brevibacterium ammoniagenes 19350
    Brevibacterium ammoniagenes 19351
    Brevibacterium ammoniagenes 19352
    Brevibacterium ammoniagenes 19353
    Brevibacterium ammoniagenes 19354
    Brevibacterium ammoniagenes 19355
    Brevibacterium ammoniagenes 19356
    Brevibacterium ammoniagenes 21055
    Brevibacterium ammoniagenes 21077
    Brevibacterium ammoniagenes 21553
    Brevibacterium ammoniagenes 21580
    Brevibacterium ammoniagenes 39101
    Brevibacterium butanicum 21196
    Brevibacterium divaricatum 21792 P928
    Brevibacterium flavum 21474
    Brevibacterium flavum 21129
    Brevibacterium flavum 21518
    Brevibacterium flavum B11474
    Brevibacterium flavum B11472
    Brevibacterium flavum 21127
    Brevibacterium flavum 21128
    Brevibacterium flavum 21427
    Brevibacterium flavum 21475
    Brevibacterium flavum 21517
    Brevibacterium flavum 21528
    Brevibacterium flavum 21529
    Brevibacterium flavum B11477
    Brevibacterium flavum B11478
    Brevibacterium flavum 21127
    Brevibacterium flavum B11474
    Brevibacterium healii 15527
    Brevibacterium ketoglutamicum 21004
    Brevibacterium ketoglutamicum 21089
    Brevibacterium ketosoreductum 21914
    Brevibacterium lactofermentum 70
    Brevibacterium lactofermentum 74
    Brevibacterium lactofermentum 77
    Brevibacterium lactofermentum 21798
    Brevibacterium lactofermentum 21799
    Brevibacterium lactofermentum 21800
    Brevibacterium lactofermentum 21801
    Brevibacterium lactofermentum B11470
    Brevibacterium lactofermentum B11471
    Brevibacterium lactofermentum 21086
    Brevibacterium lactofermentum 21420
    Brevibacterium lactofermentum 21086
    Brevibacterium lactofermentum 31269
    Brevibacterium linens 9174
    Brevibacterium linens 19391
    Brevibacterium linens 8377
    Brevibacterium paraffinolyticum 11160
    Brevibacterium spec. 717.73
    Brevibacterium spec. 717.73
    Brevibacterium spec. 14604
    Brevibacterium spec. 21860
    Brevibacterium spec. 21864
    Brevibacterium spec. 21865
    Brevibacterium spec. 21866
    Brevibacterium spec. 19240
    Corynebacterium acetoacidophilum 21476
    Corynebacterium acetoacidophilum 13870
    Corynebacterium acetoglutamicum B11473
    Corynebacterium acetoglutamicum B11475
    Corynebacterium acetoglutamicum 15806
    Corynebacterium acetoglutamicum 21491
    Corynebacterium acetoglutamicum 31270
    Corynebacterium acetophilum B3671
    Corynebacterium ammoniagenes 6872 2399
    Corynebacterium ammoniagenes 15511
    Corynebacterium fujiokense 21496
    Corynebacterium glutamicum 14067
    Corynebacterium glutamicum 39137
    Corynebacterium glutamicum 21254
    Corynebacterium glutamicum 21255
    Corynebacterium glutamicum 31830
    Corynebacterium glutamicum 13032
    Corynebacterium glutamicum 14305
    Corynebacterium glutamicum 15455
    Corynebacterium glutamicum 13058
    Corynebacterium glutamicum 13059
    Corynebacterium glutamicum 13060
    Corynebacterium glutamicum 21492
    Corynebacterium glutamicum 21513
    Corynebacterium glutamicum 21526
    Corynebacterium glutamicum 21543
    Corynebacterium glutamicum 13287
    Corynebacterium glutamicum 21851
    Corynebacterium glutamicum 21253
    Corynebacterium glutamicum 21514
    Corynebacterium glutamicum 21516
    Corynebacterium glutamicum 21299
    Corynebacterium glutamicum 21300
    Corynebacterium glutamicum 39684
    Corynebacterium glutamicum 21488
    Corynebacterium glutamicum 21649
    Corynebacterium glutamicum 21650
    Corynebacterium glutamicum 19223
    Corynebacterium glutamicum 13869
    Corynebacterium glutamicum 21157
    Corynebacterium glutamicum 21158
    Corynebacterium glutamicum 21159
    Corynebacterium glutamicum 21355
    Corynebacterium glutamicum 31808
    Corynebacterium glutamicum 21674
    Corynebacterium glutamicum 21562
    Corynebacterium glutamicum 21563
    Corynebacterium glutamicum 21564
    Corynebacterium glutamicum 21565
    Corynebacterium glutamicum 21566
    Corynebacterium glutamicum 21567
    Corynebacterium glutamicum 21568
    Corynebacterium glutamicum 21569
    Corynebacterium glutamicum 21570
    Corynebacterium glutamicum 21571
    Corynebacterium glutamicum 21572
    Corynebacterium glutamicum 21573
    Corynebacterium glutamicum 21579
    Corynebacterium glutamicum 19049
    Corynebacterium glutamicum 19050
    Corynebacterium glutamicum 19051
    Corynebacterium glutamicum 19052
    Corynebacterium glutamicum 19053
    Corynebacterium glutamicum 19054
    Corynebacterium glutamicum 19055
    Corynebacterium glutamicum 19056
    Corynebacterium glutamicum 19057
    Corynebacterium glutamicum 19058
    Corynebacterium glutamicum 19059
    Corynebacterium glutamicum 19060
    Corynebacterium glutamicum 19185
    Corynebacterium glutamicum 13286
    Corynebacterium glutamicum 21515
    Corynebacterium glutamicum 21527
    Corynebacterium glutamicum 21544
    Corynebacterium glutamicum 21492
    Corynebacterium glutamicum B8183
    Corynebacterium glutamicum B8182
    Corynebacterium glutamicum B12416
    Corynebacterium glutamicum B12417
    Corynebacterium glutamicum B12418
    Corynebacterium glutamicum B11476
    Corynebacterium glutamicum 21608
    Corynebacterium lilium P973
    Corynebacterium nitrilophilus 21419 11594
    Corynebacterium spec. P4445
    Corynebacterium spec. P4446
    Corynebacterium spec. 31088
    Corynebacterium spec. 31089
    Corynebacterium spec. 31090
    Corynebacterium spec. 31090
    Corynebacterium spec. 31090
    Corynebacterium spec. 15954 20145
    Corynebacterium spec. 21857
    Corynebacterium spec. 21862
    Corynebacterium spec. 21863

    ATCC: American Type Culture Collection, Rockville, MD, USA

    FERM: Fermentation Research Institute, Chiba, Japan

    NRRL: ARS Culture Collection, Northern Regional Research Laboratory, Peoria, IL, USA

    CECT: Coleccion Espanola de Cultivos Tipo, Valencia, Spain

    NCIMB: National Collection of Industrial and Marine Bacteria Ltd., Aberdeen, UK

    CBS: Centraalbureau voor Schimmelcultures, Baarn, NL

    NCTC: National Collection of Type Cultures, London, UK

    DSMZ: Deutsche Sammlung von Mikroorganismen und Zellkulturen, Braunschweig, Germany

    For reference see Sugawara, H. et al. (1993) World directory of collections of cultures of microorganisms: Bacteria, fungi and yeasts (4th edn), World federation for culture collections world data center on microorganisms, Saimata, Japen.
  • TABLE 4
    ALIGNMENT RESULTS
    length % homology Date of
    ID # (NT) Genbank Hit Length Accession Name of Genbank Hit Source of Genbank Hit (GAP) Deposit
    rxa00004 594 GB_IN1:CELT27F7 34660 U58762 Caenorhabditis elegans cosmid T27F7. Caenorhabditis elegans 36,442 24-MAY-1996
    GB_PR4:AC005531 161910 AC005531 Homo sapiens PAC clone DJ0701O16 from 7q33-q36, complete sequence. Homo sapiens 36,672 13-Jan-99
    GB_EST36:AV186136 360 AV186136 AV186136 Yuji Kohara unpublished cDNA: Strain N2 hermaphrodite embryo Caenorhabditis elegans 44,380 22-Jul-99
    Caenorhabditis elegans cDNA clone yk495f12 5′, mRNA sequence.
    rxa00006 558 GB_BA1:AB024708 8734 AB024708 Corynebacterium glutamicum gltB and gltD genes for glutamine 2-oxoglutarate Corynebacterium glutamicum 39,525 13-MAR-1999
    aminotransferase large and small subunits, complete cds.
    GB_EST5:N23892 434 N23892 yw46f12.s1 Weizmann Olfactory Epithelium Homo sapiens cDNA clone Homo sapiens 38,462 28-DEC-1995
    IMAGE: 255311 3′, mRNA sequence.
    GB_BA1:AB024708 8734 AB024708 Corynebacterium glutamicum gltB and gltD genes for glutamine 2-oxoglutarate Corynebacterium glutamicum 38,961 13-MAR-1999
    aminotransferase large and small subunits, complete cds.
    rxa00029
    rxa00126
    rxa00129 1620 GB_BA1:MTY20B11 36330 Z95121 Mycobacterium tuberculosis H37Rv complete genome; segment 139/162. Mycobacterium tuberculosis 40,788 17-Jun-98
    GB_BA1:MTU14909 1799 U14909 Mycobacterium tuberculosis MtrB (mtrB) gene, complete cds. Mycobacterium tuberculosis 54,422 11-Sep-96
    GB_HTG2:AC006888 140702 AC006888 Caenorhabditis elegans clone Y61A9L, ***SEQUENCING IN PROGRESS***, Caenorhabditis elegans 35,883 26-Feb-99
    2 unordered pieces.
    rxa00130 801 GB_BA1:MTY20B11 36330 Z95121 Mycobacterium tuberculosis H37Rv complete genome; segment 139/162. Mycobacterium tuberculosis 41,069 17-Jun-98
    GB_BA1:MTU01971 689 U01971 Mycobacterium tuberculosis H37Rv MtrA (mtrA) gene, complete cds. Mycobacterium tuberculosis 66,183 11-Sep-96
    GB_BA1:NMOMPR 618 X92405 N. meningitidis ompR gene. Neisseria meningitidis 50,249 31-OCT-1995
    rxa00182 3225 GB_BA1:CGPUTP 3791 Y09163 C. glutamicum putP gene. Corynebacterium glutamicum 41,126 8-Sep-97
    GB_BA1:MTV020 5143 AL021924 Mycobacterium tuberculosis H37Rv complete genome; segment 94/162. Mycobacterium tuberculosis 48,140 17-Jun-98
    GB_BA1:BSUB0019 212610 Z99122 Bacillus subtilis complete genome (section 19 of 21): from 3597091to 3809700. Bacillus subtilis 44,221 24-Jun-99
    rxa00221 342 GB_PL2:AF020584 1415 AF020584 Welwitschia mirabilis cytochrome c oxidase (coxl) gene, mitochondrial gene Mitochondrion Welwitschia 36,656 5-Jan-99
    encoding mitochondrial protein, partial cds. mirabilis
    GB_PR4:AC007421 95240 AC007421 Homo sapiens chromosome 17, clone hRPC.1030_O_14, complete sequence. Homo sapiens 35,061 27-Aug-99
    GB_BA2:AE001272 60232 AE001272 Lactococcus lactis DPC3147 plasmid pMRC01, complete plasmid sequence. Lactococcus lactis 37,764 11-Sep-98
    rxa00253 861 GB_BA2:AF126953 1638 AF126953 Corynebacterium glutamicum cystathionine gamma-synthase (metB) gene, Corynebacterium glutamicum 41,107 10-Sep-99
    complete cds.
    GB_PR3:HSJ659F15 148440 AL096791 Human DNA sequence from clone 659F15 on chromosome Xp11.21-11.4, Homo sapiens 36,190 23-Nov-99
    complete sequence.
    GB_HTG1:HS510D11 129149 Z98044 Homo sapiens chromosome 1 clone RP3-510D11, ***SEQUENCING IN Homo sapiens 36,450 23-Nov-99
    PROGRESS***, in unordered pieces.
    rxa00284 1188 GB_PR2:HS179P9 108260 Z98880 Human DNA sequence from PAC 179P9 on chromosome 6q22. Contains Homo sapiens 38,370 23-Nov-99
    transmembrane tyrosine-specific protein kinase (ROS1), ESTs and
    STS.
    GB_PR4:AF109076 113345 AF109076 Homo sapiens chromosome 7 map 7q36 BAC H6, complete sequence. Homo sapiens 35,340 13-DEC-1998
    GB_PR2:HS179P9 108260 Z98880 Human DNA sequence from PAC 179P9 on chromosome 6q22. Contains Homo sapiens 35,344 23-Nov-99
    transmembrane tyrosine-specific protein kinase (ROS1), ESTs and STS.
    rxa00287 597 GB_IN2:AF144549 7887 AF144549 Aedes albopictus ribosomal protein L34 (rpl34) gene, complete cds. Aedes albopictus 39,828 3-Jun-99
    GB_EST15:AA475366 503 AA475366 vh14e09.r1 Soares mouse mammary gland NbMMG Mus musculus cDNA Mus musculus 37,063 18-Jun-97
    clone IMAGE: 875464 5′ similar to gb: X87671 M. musculus mRNA for
    3BP-1, an SH3 domain binding (MOUSE);, mRNA sequence.
    GB_RO:MM3BP1 2359 X87671 M. musculus mRNA for 3BP-1, an SH3 domain binding protein. Mus musculus 34,635 20-OCT-1995
    rxa00291 1606 GB_PR4:AC004967 138107 AC004967 Homo sapiens clone DJ1111F22, complete sequence. Homo sapiens 36,785 5-Jun-99
    GB_EST1:M89319 418 M89319 CEL21A4 Chris Martin sorted cDNA library Caenorhabditis elegans cDNA clone Caenorhabditis elegans 38,418 02-DEC-1992
    cm21a4 5′ similar to pepsinogen A homologous peptide, mRNA sequence.
    GB_GSS15:AQ641399 569 AQ641399 RPCI93-Dpnll-28C1.TV RPCI93-Dpnll Trypanosoma brucei genomic clone Trypanosoma brucei 39,106 8-Jul-99
    RPCI93-Dpnll-28C1, genomic survey sequence.
    rxa00292 777 GB_PL1:YSCKGD2 2112 M34531 S. cerevisiae dihydrolipoyl transsuccinylase (KGD2) gene, complete cds. Saccharomyces cerevisiae 37,330 27-Apr-93
    GB_PL1:SCNUM1 9851 X61236 S. cerevisiae NUM1 gene, involved in nuclear migration control. Saccharomyces cerevisiae 36,070 06-DEC-1991
    GB_PL1:SC8358 43468 Z50046 S. cerevisiae chromosome IV cosmid 8358. Saccharomyces cerevisiae 36,070 11-Aug-97
    rxa00319 549 GB_BA1:BACJH642 282700 D84432 Bacillus subtilis DNA, 283 Kb region containing skin element. Bacillus subtilis 43,258 6-Feb-99
    GB_BA1:BSUB0014 213420 Z99117 Bacillus subtilis complete genome(section 14 of 21): from 2599451 to 2812870. Bacillus subtilis 34,264 26-Nov-97
    GB_BA1:BSUB0014 213420 Z99117 Bacillus subtilis complete genome (section 14 of 21): from 2599451 to 2812870. Bacillus subtilis 35,622 26-Nov-97
    rxa00348 519 GB_PL2:ATAC007045 68554 AC007045 Arabidopsis thaliana chromosome II BAC F23M2 genomic sequence, complete Arabidopsis thaliana 43,513 31-MAR-1999
    sequence.
    GB_PL2:ATH133743 5777 AJ133743 Arabidopsis thaliana ttg1 gene. Arabidopsis thaliana 38,247 18-Jun-99
    GB_PL1:AB010068 74589 AB010068 Arabidopsis thaliana genomic DNA, chromosome 5, TAC clone: K18P6, Arabidopsis thaliana 34,387 20-Nov-99
    complete sequence.
    rxa00350 450 GB_PL1:SCXV55KB 54719 Z70678 S. cerevisiae chromosome XV DNA, 54.7 kb region. Saccharomyces cerevisiae 35,347 16-MAY-1997
    GB_PL1:SCYOR052C 1732 Z74960 S. cerevisiae chromosome XV reading frame ORF YOR052c. Saccharomyces cerevisiae 35,347 11-Aug-97
    GB_BA1:PSE6703 2600 AJ006703 Pseudanabaena sp gene encoding for glutamine synthetase. Pseudanabaena sp. 37,978 19-Jan-99
    rxa00363 843 GB_VI:SIVMNDGB1 9215 M27470 Simian immunodeficiency virus, complete genome. Simian immunodeficiency 35,379 13-MAR-1997
    virus
    GB_OM:BTU35642 1198 U35642 Bos taurus alpha1-microglobulin/bikunin mRNA, complete cds. Bos taurus 40,131 5-Sep-96
    GB_PL1:MDO011518 1633 AJ011518 Malus domestica acc synthase gene, exons 1-4, partial. Malus domestica 40,343 23-OCT-1998
    rxa00400 1002 GB_HTG2:AC006174 203407 AC006174 Home sapiens chromosome 10 clone CIT987SK-1057L21 map 10q25, *** Homo sapiens 38,320 09-DEC-1998
    SEQUENCING IN PROGRESS***, 6 unordered pieces.
    GB_HTG2:AC006174 203407 AC006174 Homo sapiens chromosome 10 clone CIT987SK-1057L21 map 10q25, Homo sapiens 38,320 09-DEC-1998
    ***SEQUENCING IN PROGRESS***, 6 unordered pieces.
    GB_HTG2:AC006174 203407 AC006174 Home sapiens chromosome 10 clone CIT9875K-1057L21 map 10q25, Homo sapiens 37,693 09-DEC-1998
    ***SEQUENCING IN PROGRESS***, 6 unordered pieces.
    rxa00464
    rxa00494 420 GB_BA2:AF004835 40897 AF004835 Brevibacillus brevis tyrocidine biosynthesis operon, tyrocidine synthetase 1 Brevibacillus brevis 40,500 18-Nov-97
    (tycA), tyrocidine synthetase 2 (tycB), tyrocidine synthetase 3 (tycC), putative
    ABC-transporter TycD (tycD), putative ABC-transporter TycE (tycE) and
    putative thioesterase GrsT homolog (tycF) genes, complete cds.
    GB_PR3:HS84F12 78011 AL008712 Human DNA sequence from PAC 84F12 on chromosome Xq25-Xq26.3. Homo sapiens 35,749 23-Nov-99
    Contains glypican-3 precursor (intestinal protein OCI-5) (GTR2-2), ESTs and
    CA repeat.
    GB_PR3:AC005239 37005 AC005239 Homo sapiens chromosome 19, cosmid F23149, complete sequence. Homo sapiens 33,663 3-Jul-98
    rxa00516 843 GB_PR3:AF020503 206880 AF020503 Homo sapiens FRA3B common fragile region, diadenosine triphosphate Homo sapiens 40,503 23-Jan-98
    hydrolase (FHIT) gene, exon 5.
    GB_HTG2:AC007100 210344 AC007100 Homo sapiens clone NH0462D13, ***SEQUENCING IN PROGRESS***, 5 Homo sapiens 37,226 7-Apr-99
    unordered pieces.
    GB_HTG2:AC007100 210344 AC007100 Homo sapiens clone NH0462D13, ***SEQUENCING IN PROGRESS***, 5 Homo sapiens 37,226 7-Apr-99
    unordered pieces.
    rxa00551 594 GB_EST27:Al405761 607 Al405761 GH25883.5prime GH Drosophila melanogaster head pOT2 Drosophila Drosophila melanogaster 40,481 8-Feb-99
    melanogaster cDNA clone GH25883 5prime, mRNA sequence.
    GB_EST27:Al405774 607 Al405774 GH25902.5prime GH Drosophila melanogaster head pOT2 Drosophila Drosophila melanogaster 40,481 8-Feb-99
    melanogaster cDNA clone GH25902 5prime, mRNA sequence.
    GB_EST22:Al063444 674 Al063444 GH03263.5prime GH Drosophila melanogaster head pOT2 Drosophila Drosophila melanogaster 40,437 24-Nov-98
    melanogaster cDNA clone GH03263 5prime, mRNA sequence.
    rxa00583 861 GB_BA1:CORAHPS 2570 L07603 Corynebacterium glutamicum 3-deoxy-D-arabinoheptulosonate-7-phosphate Corynebacterium glutamicum 97,310 26-Apr-93
    synthase gene, complete cds.
    GB_BA1:MTV017 67200 AL021897 Mycobacterium tuberculosis H37Rv complete genome; segment 48/162. Mycobacterium tuberculosis 58,769 24-Jun-99
    GB_IN1:ACKRPA 849 X68555 A. californica KRP-A gene. Aplysia californica 41,417 30-Jun-98
    rxa00592 582 GB_IN2:AC005467 62091 AC005467 Drosophila melanogaster, chromosome 2R, region 48C1-48C2, P1 clone Drosophila melanogaster 33,565 12-DEC-1998
    DS00568, complete sequence.
    GB_IN2:AC005467 62091 AC005467 Drosophila melanogaster, chromosome 2R, region 48C1-48C2, P1 clone Drosophila melanogaster 35,893 12-DEC-1998
    DS00568, complete sequence.
    rxa00593 471 GB_BA1:MTV025 121125 AL022121 Mycobacterium tuberculosis H37Rv complete genome; segment 155/162. Mycobacterium tuberculosis 33,761 24-Jun-99
    GB_BA1:MSGB577COS 37770 L01263 M. leprae genomic dna sequence, cosmid b577. Mycobacterium leprae 35,065 14-Jun-96
    GB_BA2:AF114720 2366 AF114720 Xanthomonas campestris pv. vesicatoria avirulence protein AvrBs2 (avrBs2) Xanthomonas campestris pv. 37,768 1-Feb-99
    gene, complete cds. vesicatoria
    rxa00603 576 GB_BA1:RCPUTRA 4357 X78346 R. capsulatus (B10S) putR and putA genes. Rhodobacter capsulatus 34,867 08-DEC-1995
    GB_GSS10:AQ227452 474 AQ227452 HS_2015_B2_B07_MR CIT Approved Human Genomic Sperm Library D Homo sapiens 35,337 26-Sep-98
    Homo sapiens genomic clone Plate = 2015 Col = 14 Row = D,
    genomic survey sequence.
    GB_GSS3:B60643 251 B60643 CIT-HSP-2015D14.TRB CIT-HSP Homo sapiens genomic clone 2015D14, Homo sapiens 39,200 21-Jun-98
    genomic survey sequence.
    rxa00609 558 GB_HTG3:AC009346 105005 AC009346 Drosophila melanogaster chromosome 3 clone BACR03P13 (D672) RPCI-98 Drosophila melanogaster 31,261 27-Aug-99
    03.P.13 map 83A-83B strain y; cn bw sp, ***SEQUENCING IN
    PROGRESS***, 83 unordered pieces.
    GB_HTG3:AC009346 105005 AC009346 Drosophila melanogaster chromosome 3 clone BACR03P13 (D672) RPCI-98 Drosophila melanogaster 31,261 27-Aug-99
    03.P.13 map 83A-83B strain y; cn bw sp, ***SEQUENCING IN
    PROGRESS***, 83 unordered pieces.
    GB_HTG3:AC009346 105005 AC009346 Drosophila melanogaster chromosome 3 clone BACR03P13 (D672) RPCI-98 Drosophila melanogaster 30,072 27-Aug-99
    03.P.13 map 83A-83B strain y; cn bw sp, ***SEQUENCING IN
    PROGRESS***, 83 unordered pieces.
    rxa00630 828 GB_BA1:MTCY369 36850 Z80226 Mycobacterium tuberculosis H37Rv complete genome; segment 36/162. Mycobacterium tuberculosis 60,870 17-Jun-98
    GB_BA1:SC4H8 15560 AL020958 Streptomyces coelicolor cosmid 4H8. Streptomyces coelicolor 48,474 10-DEC-1997
    GB_BA1:MTCY20G9 37218 Z77162 Mycobacterium tuberculosis H37Rv complete genome; segment 25/162. Mycobacterium tuberculosis 46,537 17-Jun-98
    rxa00651 1455 GB_PR2:AP000165 100000 AP000165 Homo sapiens genomic DNA, chromosome 21q22.1, D21S226-AML region, Homo sapiens 35,685 20-Nov-99
    clone B2344F14-f50E8, segment 1/9, complete sequence.
    GB_RO:AC005835 132297 AC005835 Mus musculus clone UWGC: mbac82 from 14D1-D2 (T-Cell Receptor Alpha Mus musculus 37,851 21-OCT-1998
    Locus), complete sequence.
    GB_PR2:AP000165 100000 AP000165 Homo sapiens genomic DNA, chromosome 21q22.1, D21S226-AML region, Homo sapiens 35,610 20-Nov-99
    clone B2344F14-f50E8, segment 1/9, complete sequence.
    rxa00655 762 GB_PR3:AC004460 113803 AC004460 Homo sapiens PAC clone DJ1086D14, complete sequence. Homo sapiens 38,606 24-MAR-1998
    GB_PL1:CRERSP4A 7707 M87526 Chlamydomonas reinhardtii flagellar radial spoke protein (RSP4) and RSP6) Chlamydomonas reinhardtii 39,067 27-Apr-93
    genes, complete cds.
    GB_EST38:AW041495 517 AW041495 EST284359 tomato mixed elicitor, BTI Lycopersicon esculentum cDNA clone Lycopersicon esculentum 38,760 18-OCT-1999
    cLET14F2, mRNA sequence.
    rxa00813 1254 GB_BA1:MSGMPB70B 1009 D38230 Mycobacterium bovis DNA for MPB70, complete cds, strain: BCG Tokyo. Mycobacterium bovis 40,956 8-Feb-99
    GB_BA1:MTCY274 39991 Z74024 Mycobacterium tuberculosis H37Rv complete genome; segment 126/162. Mycobacterium tuberculosis 41,447 19-Jun-98
    GB_BA1:MSGMPB70A 1009 D38229 Mycobacterium bovis DNA for MPB70, complete cds, strain: BCG Pasteur. Mycobacterium bovis 40,956 8-Feb-99
    rxa00822 804 GB_BA1:MTV025 121125 AL022121 Mycobacterium tuberculosis H37Rv complete genome; segment 155/162. Mycobacterium tuberculosis 64,925 24-Jun-99
    GB_EST35:AI857185 646 AI857185 603007G10.x1 603 —stressed root cDNA library from Wang/Bohnert lab Zea Zea mays 40,206 16-Jul-99
    mays cDNA, mRNA sequence.
    GB_PR3:HS95C20 138849 Z97181 Homo sapiens DNA sequence from PAC 95C20 on chromosome Xp11.3-11.4. Homo sapiens 37,633 23-Nov-99
    Contains STSs and the DXS7 locus with GT and GTG repeat polymorphisms,
    complete sequence.
    rxa00848 2043 GB_BA1:MTCI65 34331 Z95584 Mycobacterium tuberculosis H37Rv complete genome; segment 50/162. Mycobacterium tuberculosis 63,215 17-Jun-98
    GB_BA1:MSGY348 40056 AD000020 Mycobacterium tuberculosis sequence from clone y348. Mycobacterium tuberculosis 47,938 10-DEC-1996
    GB_HTG3:AC008608 207341 AC008608 Homo sapiens chromosome 5 clone CIT978SKB_113I20, ***SEQUENCING IN Homo sapiens 43,001 3-Aug-99
    rxa00849 444 GB_HTG4:AC007305 216524 AC007305 Mus musculus, ***SEQUENCING IN PROGRESS***, 10 unordered pieces. Mus musculus 38,979 23-OCT-1999
    GB_HTG4:AC007305 216524 AC007305 Mus musculus, ***SEQUENCING IN PROGRESS***, 10 unordered pieces. Mus musculus 38,979 23-OCT-1999
    GB_HTG4:AC007305 216524 AC007305 Mus musculus, ***SEQUENCING IN PROGRESS***, 10 unordered pieces. Mus musculus 36,636 23-OCT-1999
    rxa00885 1149 GB_EST36:AV178106 300 AV178106 AV178106 Yuji Kohara unpublished cDNA: Strain N2 hermaphrodite embryo Caenorhabditis elegans 39,057 21-Jul-99
    Caenorhabditis elegans cDNA clone yk538b7 3′, mRNA sequence.
    GB_EST16:C30090 300 C30090 C30090 Yuji Kohara unpublished cDNA: Strain N2 hermaphrodite embryo Caenorhabditis elegans 38,000 18-OCT-1999
    Caenorhabditis elegans cDNA clone yk236d2 3′, mRNA sequence.
    GB_IN1:CET20D3 32679 Z68220 Caenorhabditis elegans cosmid T20D3, complete sequence. Caenorhabditis elegans 36,067 2-Sep-99
    rxa00894 1251 GB_EST20:AA890839 281 AA890839 TENS0689 T. cruzi epimastigote normalized cDNA Library Trypanosoma cruzi Trypanosoma cruzi 39,779 29-OCT-1998
    cDNA clone 689 5′, mRNA sequence.
    GB_EST20:AA890838 284 AA890838 TENS0687 T. cruzi epimastigote normalized cDNA Library Trypanosoma cruzi Trypanosoma cruzi 39,674 29-OCT-1998
    cDNA clone 687 5′, mRNA sequence.
    GB_RO:RNMAFAEX2 1709 X97192 R. norvegicus MAFA gene, exon2. Rattus norvegicus 36,989 17-Apr-96
    rxa00947 459 GB_EST6:W04640 420 W04640 zb93b03.s1 Soares_parathyroid_tumor_NbHPA Homo sapiens cDNA clone Homo sapiens 43,519 23-Apr-96
    IMAGE:320333 3′, mRNA sequence.
    GB_EST6:W04640 420 W04640 zb93b03.s1 Soares_parathyroid_tumor_NbHPA Homo sapiens cDNA clone Homo sapiens 37,725 23-Apr-96
    IMAGE:320333 3′, mRNA sequence.
    rxa01001
    rxa01065 1038 GB_BA1:MTCY27 27548 Z95208 Mycobacterium tuberculosis H37Rv complete genome; segment 104/162. Mycobacterium tuberculosis 38,949 17-Jun-98
    GB_BA2:AF065159 35209 AF065159 Bradyrhizobium japonicum putative arylsulfatase (arsA), putative soluble lytic Bradyrhizobium japonicum 46,369 27-OCT-1999
    transglycosylase precursor (sltA), dihydrodipicolinate synthase (dapA), MscL
    GB_HTG2:AC006794 297866 AC006794 Caenorhabditis elegans clone Y50D4a, ***SEQUENCING IN PROGRESS***, Caenorhabditis elegans 34,676 23-Feb-99
    29 unordered pieces.
    rxa01110 696 GB_HTG7:AC009530 204901 AC009530 Homo sapiens chromosome 7, ***SEQUENCING IN PROGRESS***, 32 Homo sapiens 36,364 08-DEC-1999
    unordered pieces.
    GB_HTG3:AC009301 163369 AC009301 Homo sapiens clone NH0062F14, ***SEQUENCING IN PROGRESS***, 5 Homo sapiens 34,538 13-Aug-99
    unordered pieces.
    GB_HTG3:AC009301 163369 AC009301 Homo sapiens clone NH0062F14, ***SEQUENCING IN PROGRESS***, 5 Homo sapiens 34,538 13-Aug-99
    unordered pieces.
    rxa01118 888 GB_BA2:AF003947 5475 AF003947 Rhodococcus opacus succinyl CoA:3-oxoadipate CoA transferase subunit Rhodococcus opacus 55,982 12-MAR-1998
    homolog (pcal') gene, partial cds, protocatechuate dioxygenase beta subunit
    (pcaH), protocatechuate dioxygenase alpha subunit (pcaG), 3-carboxy-
    cis,cis-muconate cycloisomerase homolog (pcaB), 3-oxoadipate enol-lactone
    hydrolase/4-carboxymuconolactone decarboxylase (pcaL) and PcaR (pcaR)
    genes, complete cds, and 3-oxoadipyl CoA thiolase homolog (pcaF') gene,
    partial cds.
    GB_BA1:ROX99622 7224 X99622 Rhodococcus opacus catR, catA, catB, catC genes and five ORFs. Rhodococcus opacus 40,000 24-Sep-97
    GB_IN1:CELC14F5 42966 U29082 Caenorhabditis elegans cosmid C14F5. Caenorhabditis elegans 37,485 15-Jun-95
    rxa01125 336 GB_EST16:C41499 360 C41499 C41499 Yuji Kohara unpublished cDNA:Strain N2 hermaphrodite embryo Caenorhabditis elegans 44,747 18-OCT-1999
    Caenorhabditis elegans cDNA clone yk268f1 5′, mRNA sequence.
    GB_HTG2:AC006705 195349 AC006705 Caenorhabditis elegans clone Y108G3c, ***SEQUENCING IN PROGRESS***, Caenorhabditis elegans 42,415 23-Feb-99
    2 unordered pieces.
    GB_IN2:CELF33E11 36400 AF067622 Caenorhabditis elegans cosmid F33E11. Caenorhabditis elegans 42,415 27-MAY-1999
    rxa01211 1380 GB_EST28:AI520492 503 AI520492 LD40669.3prime LD Drosophila melanogaster embryo pOT2 Drosophila Drosophila melanogaster 40,726 16-MAR-1999
    melanogaster cDNA clone LD40669 3prime, mRNA sequence.
    GB_EST27:AI403753 551 AI403753 GH23256.3prime GH Drosophila melanogaster head pOT2 Drosophila Drosophila melanogaster 41,316 8-Feb-99
    melanogaster cDNA clone GH23256 3prime, mRNA sequence.
    GB_EST19:AA391230 493 AA391230 LD10605.3prime LD Drosophila melanogaster embryo BlueScript Drosophila Drosophila melanogaster 38,415 27-Nov-98
    melanogaster cDNA clone LD10605 3prime, mRNA sequence.
    rxa01241 603 GB_BA1:U00019 36033 U00019 Mycobacterium leprae cosmid B2235. Mycobacterium leprae 58,783 01-MAR-1994
    GB_BA1:MSGB42CS 22781 L78826 Mycobacterium leprae cosmid B42 DNA sequence. Mycobacterium leprae 58,464 15-Jun-96
    GB_HTG5:AC007521 173897 AC007521 Drosophila melanogaster chromosome X clone BACR49A04 (D698) RPCI-98 Drosophila melanogaster 40,137 17-Nov-99
    49.A.4 map 10A2-10B2 strain y; cn bw sp, ***SEQUENCING IN
    PROGRESS***, 56 unordered pieces.
    rx01248 529 GB_BA1:ECOUW93 338534 U14003 Escherichia coli K-12 chromosomal region from 92.8 to 00.1 minutes. Escherichia coli 40,546 17-Apr-96
    GB_BA1:D90900 137740 D90900 Synechocystis sp. PCC6803 complete genome, 2/27, 133860-271599. Synechocystis sp. 32,177 7-Feb-99
    GB_BA1:ECOUW93 338534 U14003 Escherichia coli K-12 chromosomal region from 92.8 to 00.1 minutes. Escherichia coli 37,044 17-Apr-96
    rxa01272 726 GB_EST10:AA181367 520 AA181367 zp42c11.s1 Stratagene muscle 937209 Homo sapiens cDNA clone Homo sapiens 41,408 09-MAR-1998
    IMAGE: 612116 3′, mRNA sequence.
    GB_VI:PBU42580 330742 U42580 Paramecium bursaria Chlorella virus 1, complete genome. Paramecium bursaria 38,265 4-Nov-99
    Chlorella virus 1
    GB_VI:AF063866 236120 AF063866 Melanoplus sanguinipes entomopoxvirus, complete genome. Melanoplus sanguinipes 38,579 22-DEC-1998
    entomopoxvirus
    rxa01368 435 GB_BA2:AF164439 783 AF164439 Mycobacterium smegmatis WhmD (whmD) gene, complete cds; and unknown Mycobacterium smegmatis 57,477 4-Aug-99
    gene.
    GB_BA1:MTV015 1668 AL021840 Mycobacterium tuberculosis H37Rv complete genome; segment 140/162. Mycobacterium tuberculosis 37,617 17-Jun-98
    GB_BA1:SGWHIB 593 X68708 S. griseocarneum whiB-Stv gene. Streptomyces griseocarneus 53,396 17-Jan-94
    rxa01375 1578 GB_BA1:MTCY71 42729 Z92771 Mycobacterium tuberculosis H37Rv complete genome; segment 141/162. Mycobacterium tuberculosis 52,638 10-Feb-99
    GB_IN2:AC005935 29330 AC005935 Leishmania major chromosome 3 clone L7234 strain Friedlin, complete Leishmania major 39,777 15-Nov-99
    sequence.
    GB_IN2:AF005195 1962 AF005195 Trypanosoma cruzi paraflagellar rod component Par3 (par3b) mRNA, complete Trypanosoma cruzi 40,304 17-Aug-98
    cds.
    rxa01418 369 GB_IN2:CELC53B7 29535 U42830 Caenorhabditis elegans cosmid C53B7. Caenorhabditis elegans 34,375 03-MAR-1998
    GB_IN1:CEU49449 1118 U49449 Caenorhabditis elegans olfactory receptor Odr-10 (odr-10) mRNA, complete Caenorhabditis elegans 47,111 17-MAY-1996
    cds.
    GB_EST35:AI871077 295 AI871077 wl70c12.x1 NCI_CGAP_Brn25 Homo sapiens cDNA clone IMAGE: 2430262 3′ Homo sapiens 37,722 30-Aug-99
    similar to gb: X70683 cds1 SOX-4 PROTEIN (HUMAN);, mRNA sequence.
    rxa01450 687 GB_BA1:MTV017 67200 AL021897 Mycobacterium tuberculosis H37Rv complete genome; segment 48/162. Mycobacterium tuberculosis 60,059 24-Jun-99
    GB_BA1 MAMAMIRM 4972 X79027 M. ammoniaphilum genes mamIR and mamIM. Microbacterium 39,912 20-Nov-96
    ammoniaphilum
    GB_HTG3:AC009121 46469 AC009121 Homo sapiens chromosome 16 clone RPCI-11_485G7, ***SEQUENCING IN Homo sapiens 55,507 3-Aug-99
    PROGRESS***, 32 unordered pieces.
    rxa01451 690 GB_BA1:MTV017 67200 AL021897 Mycobacterium tuberculosis H37Rv complete genome; segment 48/162. Mycobacterium tuberculosis 63,516 24-Jun-99
    GB_BA1:MAMAMIRM 4972 X79027 M. ammoniaphilum genes mamIR and mamIM, Microbacterium 37,113 20-Nov-96
    ammoniaphilum
    GB_BA1:MLCB1222 34714 AL049491 Mycobacterium leprae cosmid B1222. Mycobacterium leprae 36,324 27-Aug-99
    rxa01500 567 GB_IN1:CEC09G5 29688 Z46791 Caenorhabditis elegans cosmid C09G5, complete sequence. Caenorhabditis elegans 36,298 2-Sep-99
    GB_GSS9:AQ096256 390 AQ096256 HS_3037_A1_F11_MF CIT Approved Human Genomic Sperm Library D Homo sapiens 46,316 27-Aug-98
    Homo sapiens genomic clone Plate = 3037 Col = 21 Row = K,
    genomic survey sequence.
    GB_HTG1:HS1099D15 1301 AL035456 Homo sapiens chromosome 20 clone RP5-1099D15, ***SEQUENCING IN Homo sapiens 39,388 23-Nov-99
    PROGRESS***, in unordered pieces.
    rxa01537 774 GB_RO:RNCYCBMR 2354 X64589 R. norvegicus mRNA for cyclin B. Rattus norvegicus 40,584 29-MAR-1994
    GB_RO:RATCYCLINB 1465 L11995 Rattus norvegicus cyclin B mRNA, complete cds. Rattus norvegicus 40,584 3-Feb-98
    GB_RO:RNCYCLNB 1902 X60768 Rat mRNA for cyclin B. Rattus norvegicus 40,530 15-Aug-96
    rxa01573 2205 GB_HTG4:AC011317 40524 AC011317 Homo sapiens chromosome 3 seeders clone RPCI11-103G8, Homo sapiens 34,814 21-OCT-1999
    ***SEQUENCING IN PROGRESS***, 31 unordered pieces.
    GB_HTG4:AC011317 40524 AC011317 Homo sapiens chromosome 3 seeders clone RPCI11-103G8, Homo sapiens 34,814 21-OCT-1999
    ***SEQUENCING IN PROGRESS***, 31 unordered pieces.
    GB_IN1:CELK06A5 24323 AF039038 Caenorhabditis elegans cosmid K06A5. Caenorhabditis elegans 38,899 1-Jan-98
    rxa01655 1482 GB_GSS15:AQ624398 460 AQ624398 HS_2106_B2_C03_T7C CIT Approved Human Genomic Sperm Library D Homo sapiens 36,449 16-Jun-99
    Homo sapiens genomic clone Plate = 2106 Col = 6 Row = F, genomic survey
    sequence.
    GB_BA1:SC6G10 36734 AL049497 Streptomyces coelicolor cosmid 6G10. Streptomyces coelicolor 39,098 24-MAR-1999
    rxa01687 GB_BA1:MLCB268 38859 AL022602 Mycobacterium leprae cosmid B268. Mycobacterium leprae 39,891 27-Aug-99
    rxa01759 885 GB_OV:PMU11880 16201 U11880 Petromyzon marinus mitochondrion, complete genome. Mitochondrion Petromyzon 36,977 24-Sep-96
    marinus
    GB_STS:G39160 605 G39160 Z13915 Zebrafish AB Danio rerio STS genomic, sequence tagged site. Danio rerio 36,093 30-Jul-98
    GB_STS:G39160 605 G39160 Z13915 Zebrafish AB Danio rerio STS genomic, sequence tagged site. Danio rerio 36,093 30-Jul-98
    rxa01763 588 GB_GSS4:AQ701186 454 AQ701186 HS_2129_A2_D04_T7C CIT Approved Human Genomic Sperm Library D Homo sapiens 40,000 7-Jul-99
    Homo sapiens genomic clone Plate = 2129 Col = 8 Row = G, genomic survey
    sequence.
    GB_BA1:ENEPPD1 5363 D28859 Enterococcus faecalis Plasmid pPD1 DNA for iPD1, TraB, TraA, ORF1and Enterococcus faecalis 37,117 7-Feb-99
    TraC, complete cds.
    GB_BA1:ENEPPD1A 8526 D78016 Enterococcus faecalis Plasmid pPD1 genes for REPB, REPA, TRAC, TRAB, Enterococcus faecalis 35,788 5-Feb-99
    TRAA, iPD1, TRAE, TRAF, complete cds and partial cds.
    rxa01826 2061 GB_BA1:MLCB1770 37821 Z70722 Mycobacterium leprae cosmid B1770. Mycobacterium leprae 37,524 29-Aug-97
    GB_BA1:SCH69 35824 AL079308 Streptomyces coelicolor cosmid H69. Streptomyces coelicolor 51,185 15-Jun-99
    GB_BA1:SCGD3 33779 AL096822 Streptomyces coelicolor cosmid GD3. Streptomyces coelicolor 38,775 8-Jul-99
    rxa01827 1530 GB_BA1:MTCY10H4 39160 Z80233 Mycobacterium tuberculosis H37Rv complete genome; segment 2/162. Mycobacterium tuberculosis 37,815 17-Jun-98
    GB_BA1:AB016932 2711 AB016932 Streptomyces coelicolor gene for protein serine/threonine kinase, complete cds. Streptomyces coelicolor 42,543 11-Nov-98
    GB_RO:AF145705 2201 AF145705 Mus musculus T2K protein kinase homolog mRNA, complete cds. Mus musculus 40,438 2-Jun-99
    rxa01830 1476 GB_PR2:HSU82672 156854 U82672 Human chromosome X clone Qc15B1, complete sequence. Homo sapiens 36,389 12-MAY-1997
    GB_BA2:AF087482 26245 AF087482 Pseudomonas aeruginosa clcC and ohbH genes, Lys-R type regulatory protein Pseudomonas aeruginosa 40,805 31-OCT-1998
    (clcR), chlorocatechol-1,2-dioxygenase (clcA), chloromuconate cycloisomerase
    (clcB), dienelactone hydrolase (clcD), maleylacetate reductase (clcE),
    transposase (tnpA), ATP-binding protein (tnpB), putative regulatory protein
    (ohbR), o-halobenzoate dioxygenase reductase (ohbA), o-halobenzoate
    dioxygenase alpha subunit (ohbB), o-halobenzoate dioxygenase beta subunit
    (ohbC), o-halobenzoate dioxygenase ferredoxin (ohbD), putative membrane
    spanning protein (ohbE), ATP-binding protein (ohbF), putative substrate binding
    protein (ohbG), and putative dioxygenase genes, complete cds; and unknown
    gene.
    GB_PR2:HSU82672 156854 U82672 Human chromosome X clone Qc15B1, complete sequence. Homo sapiens 36,301 12-MAY-1997
    rxa01836 828 GB_GSS1:CI22H2 704 AJ227010 Ciona intestinalis genomic fragment, clone 22H2, genomic survey sequence. Ciona intestinalis 33,481 10-MAR-1998
    GB_EST18:AA692868 461 AA692868 vr58h12.s1 Knowles Solter mouse 2 cell Mus musculus cDNA clone Mus musculus 47,222 16-DEC-1997
    IMAGE: 1124903 5′, mRNA sequence.
    GB_PR3:HSDJ860P4 156791 AL049594 Human DNA sequence from clone 860P4 on chromosome 20 Contains ESTs, Homo sapiens 35,504 23-Nov-99
    STSs, GSSs and a CpG island, complete sequence.
    rxa01840 654 GB_BA1:D90914 145709 D90914 Synechocystis sp. PCC6803 complete genome, 16/27, 1991550-2137258. Synechocystis sp. 61,315 7-Feb-99
    GB_EST25:AU041657 306 AU041657 AU041657 Mouse four-cell-embryo cDNA Mus musculus cDNA clone Mus musculus 39,216 04-DEC-1998
    J1007D01 3′, mRNA sequence.
    GB_PL2:AAU82633 474 U82633 Alternaria alternata Alt a I subunit mRNA, complete cds. Alternaria alternata 45,092 13-Jan-97
    rxa01860 1008 GB_PL2:AC004255 97789 AC004255 Arabidopsis thaliana BAC T1F9 chromosome 1, complete sequence. Arabidopsis thaliana 35,939 16-Apr-98
    GB_BA1:BSUB0004 213190 Z99107 Bacillus subtilis complete genome (section 4 of 21): from 600701 to 813890. Bacillus subtilis 37,111 26-Nov-97
    GB_BA1:D86418 20341 D86418 Bacillus subtilis genomic DNA 69-70 degree region, partial sequence. Bacillus subtilis 38,352 7-Feb-99
    rxa01861 2088 GB_HTG4:AC009949 173517 AC009949 Homo sapiens chromosome unknown clone NH0069J07, WORKING DRAFT Homo sapiens 36,544 29-OCT-1999
    SEQUENCE, in unordered pieces.
    GB_HTG4:AC009949 173517 AC009949 Homo sapiens chromosome unknown clone NH0069J07, WORKING DRAFT Homo sapiens 36,544 29-OCT-1999
    SEQUENCE, in unordered pieces.
    GB_HTG4:AC009949 173517 AC009949 Homo sapiens chromosome unknown clone NH0069J07, WORKING DRAFT Homo sapiens 35,676 29-OCT-1999
    SEQUENCE, in unordered pieces.
    rxa01898 816 GB_HTG1:CEY48B6 293827 AL021151 Caenorhabditis elegans chromosome II clone Y48B6, ***SEQUENCING IN Caenorhabditis elegans 33,250 1-Apr-99
    PROGRESS***, in unordered pieces.
    GB_HTG1:CEY48B6 293827 AL021151 Caenorhabditis elegans chromosome II clone Y48B6, ***SEQUENCING IN Caenorhabditis elegans 33,250 1-Apr-99
    PROGRESS***, in unordered pieces.
    GB_HTG1:CEY53F4_2 110000 Z92860 Caenorhabditis elegans chromosome II clone Y53F4, ***SEQUENCING IN Caenorhabditis elegans 34,766 Z92860
    PROGRESS***, in unordered pieces.
    rxa01935 1287 GB_PR3:HSBA259P1 48084 AL080273 Human DNA sequence from clone 259P1 on chromosome 22. Contains STSs, Homo sapiens 38,661 23-Nov-99
    GSSs, genomic markers D22S1154, D22S310 and D22S690, and a gt repeat
    polymorphism, complete sequence.
    GB_BA1:RHMIND 2862 M19019 R. fredii host-inducible protein genes A and B, complete cds. Sinorhizobium fredii 37,007 26-Apr-93
    GB_BA2:AE000108 10894 AE000108 Rhizobium sp. NGR234 plasmid pNGR234a, section 45 of 46 of the complete Rhizobium sp. NGR234 37,322 12-DEC-1997
    plasmid sequence.
    rxa02127 777 GB_BA1:D90911 143051 D90911 Synechocystis sp. PCC6803 complete genome, 13/27, 1576593-1719643. Synechocystis sp. 35,480 7-Feb-99
    GB_PR2:AC002477 124095 AC002477 Human PAC clone DJ327A19 from Xq25-q26, complete sequence. Homo sapiens 35,409 22-Aug-97
    GB_PR2:AC002477 124095 AC002477 Human PAC clone DJ327A19 from Xq25-q26, complete sequence. Homo sapiens 38,536 22-Aug-97
    rxa02210 687 GB_BA1:AB025424 2995 AB025424 Corynebacterium glutamicum gene for aconitase, partial cds. Corynebacterium glutamicum 100,000 3-Apr-99
    GB_EST15:AA534896 490 AA534896 nf78e02.s1 NCI_CGAP_Co3 Homo sapiens cDNA clone IMAGE: 926042 3′, Homo sapiens 38,929 21-Aug-97
    mRNA sequence.
    GB_BA1:AB025424 2995 AB025424 Corynebacterium glutamicum gene for aconitase, partial cds. Corynebacterium glutamicum 41,119 3-Apr-99
    rxa02232 1650 GB_BA1:MTCY154 13935 Z98209 Mycobacterium tuberculosis H37Rv complete genome; segment 121/162. Mycobacterium tuberculosis 38,882 17-Jun-98
    GB_BA1:MSGY154 40221 AD000002 Mycobacterium tuberculosis sequence from clone y154. Mycobacterium tuberculosis 56,593 03-DEC-1996
    GB_BA1:SC4H2 38400 AL022268 Streptomyces coelicolor cosmid 4H2. Streptomyces coelicolor 55,569 6-Apr-98
    rxa02270 744 GB_BA1:AP000004 217000 AP000004 Pyrococcus horikoshii OT3 genomic DNA, 777001-994000 nt. position (4/7). Pyrococcus horikoshii 36,190 8-Feb-99
    GB_BA1:AP000004 217000 AP000004 Pyrococcus horikoshii OT3 genomic DNA, 777001-994000 nt. position (4/7). Pyrococcus horikoshii 36,951 8-Feb-99
    GB_HTG3:AC008403 199233 AC008403 Homo sapiens chromosome 19 clone CIT-HSPC_273B12, ***SEQUENCING Homo sapiens 38,420 3-Aug-99
    IN PROGRESS***, 82 unorderd pieces.
    rxa02306 414 GB_EST8:AA011641 313 AA011641 zi02e11.s1 Soares_fetal_liver_spleen_1NFLS_S1 Homo sapiens cDNA clone Homo sapiens 35,235 09-MAY-1997
    IMAGE: 429644 3′, mRNA sequence.
    GB_GSS1:CNS00NAO 527 AL081678 Arabidopsis thaliana genome survey sequence SP6 end of BAC F3H19 of IGF Arabidopsis thaliana 40,615 28-Jun-99
    library from strain Columbia of Arabidopsis thaliana, genomic survey sequence.
    GB_EST24:C97772 494 C97772 C97772 Rice callus Oryza sativa cDNA clone C62702_6Z, mRNA sequence. Oryza sativa 36,667 19-OCT-1998
    rxa02365 1968 GB_BA1:U00016 42931 U00016 Mycobacterium leprae cosmid B1937. Mycobacterium leprae 67,483 01-MAR-1994
    GB_BA1:MTCY253 41230 Z81368 Mycobacterium tuberculosis H37Rv complete genome; segment 106/162. Mycobacterium tuberculosis 37,888 17-Jun-98
    GB_BA1:BACJH642 282700 D84432 Bacillus subtilis DNA, 283 Kb region containing skin element. Bacillus subtilis 58,496 6-Feb-99
    rxa02376 1626 GB_BA2:CGU31230 3005 U31230 Corynebacterium glutamicum Obg protein homolog gene, partial cds, gamma Corynebacterium glutamicum 97,504 2-Aug-96
    glutamyl kinase (proB) gene, complete cds, and (unkdh) gene, complete cds.
    GB_BA1:D87915 1647 D87915 Streptomyces coelicolor DNA for Obg, complete cds. Streptomyces coelicolor 58,013 7-Feb-99
    GB_BA1:MTV016 53662 AL021841 Mycobacterium tuberculosis H37Rv complete genome; segment 143/162. Mycobacterium tuberculosis 38,051 23-Jun-99
    rxa02450 678 GB_BA2:AE000654 12391 AE000654 Helicobacter pylori 26695 section 132 of 134 of the complete genome. Helicobacter pylori 26695 36,269 6-Apr-99
    GB_HTG3:AC009298 165826 AC009298 Homo sapiens clone NH0017106, ***SEQUENCING IN PROGRESS***, 2 Homo sapiens 35,886 13-Aug-99
    unordered pieces.
    GB_HTG4:AC010187_2 110000 AC010187 Homo sapiens chromosome 3 seeders clone RPCI11-389O9, Homo sapiens 38,939 AC010187
    ***SEQUENCING IN PROGRESS***, 164 unordered pieces.
    rxa02493 1362 GB_BA1:CGBETPGEN 2339 X93514 C. glutamicum betP gene. Corynebacterium glutamicum 38,346 8-Sep-97
    GB_BA1:SHGCPIR 107379 X86780 S. hygroscopicus gene cluster for polyketide immunosuppressant rapamycin. Streptomyces hygroscopicus 42,556 16-Aug-96
    GB_HTG2:AC007084 138793 AC007084 Drosophila melanogaster chromosome 2 clone BACR26A16 (D577) RPCI-98 Drosophila melanogaster 35,985 2-Aug-99
    26.A.16 map 43F-44A strain y; cn bw sp, ***SEQUENCING IN
    PROGRESS***, 19 unordered pieces.
    rxa02494 819 GB_BA1:U00018 42991 U00018 Mycobacterium leprae cosmid B2168. Mycobacterium leprae 42,105 01-MAR-1994
    GB_BA1:MTCY20G9 37218 Z77162 Mycobacterium tuberculosis H37Rv complete genome; segment 25/162. Mycobacterium tuberculosis 64,552 17-Jun-98
    GB_BA1:MBY13627 3208 Y13627 Mycobacterium bovis BCG senX3, regX3 genes. Mycobacterium bovis BCG 64,428 6-Jan-98
    rxa02631 1488 GB_EST17:AA655226 468 AA655226 vq84a10.s1 Knowles Solter mouse 2 cell Mus musculus cDNA clone Mus musculus 36,052 4-Nov-97
    IMAGE: 1108986 5′ similar to gb: J03827 Y BOX BINDING PROTEIN-1
    (HUMAN); gb: M62867 Mouse Y box transcription factor (MOUSE);, mRNA
    sequence.
    GB_GSS1:CNS012GD 898 AL101527 Drosophila melanogaster genome survey sequence T7 end of BAC Drosophila melanogaster 34,449 26-Jul-99
    BACN07L05 of DrosBAC library from Drosophila melanogaster (fruit fly),
    genomic survey sequence.
    GB_GSS3:B10133 1137 B10133 F2H22-T7 IGF Arabidopsis thaliana genomic clone F2H22, genomic survey Arabidopsis thaliana 38,011 14-MAY-1997
    sequence.
    rxa02632 819 GB_BA1:MTCY369 36850 Z80226 Mycobacterium tuberculosis H37Rv complete genome; segment 36/162. Mycobacterium tuberculosis 50,124 17-Jun-98
    GB_BA1:S76966 480 S76966 {BCG2 insert site} [Mycobacterium tuberculosis, BCG Japan, IS6110/IS986, Mycobacterium tuberculosis 39,437 27-Jul-95
    Insertion, 480 nt].
    GB_PR3:AC005019 188362 AC005019 Homo sapiens BAC clone GS250A16 from 7p21-p22, complete sequence. Homo sapiens 36,763 27-Aug-98
    rxa02667 717 GB_BA1:MSGY23 40806 AD000016 Mycobacterium tuberculosis sequence from clone y23. Mycobacterium tuberculosis 55,742 10-DEC-1996
    GB_BA1:MTV024 8189 AL022075 Mycobacterium tuberculosis H37Rv complete genome; segment 151/162. Mycobacterium tuberculosis 39,474 17-Jun-98
    GB_BA1:MLCB1450 38065 AL035159 Mycobacterium leprae cosmid B1450. Mycobacterium leprae 39,898 27-Aug-99
    rxa02668 846 GB_HTG2:AC007739 158262 AC007739 Homo sapiens clone NH0091L03, ***SEQUENCING IN PROGRESS***, 4 Homo sapiens 38,659 5-Jun-99
    unordered pieces.
    GB_HTG2:AC007739 158262 AC007739 Homo sapiens clone NH0091L03, ***SEQUENCING IN PROGRESS***, 4 Homo sapiens 38,659 5-Jun-99
    unordered pieces.
    GB_EST24:AI190741 443 AI190741 qd61a09.x1 Soares_testis_NHT Homo sapiens cDNA clone IMAGE: 1733944 Homo sapiens 39,661 28-OCT-1998
    3′, mRNA sequence.
    rxa02669 1239 GB_HTG2: AC007739 158262 AC007739 Homo sapiens clone NH0091L03, ***SEQUENCING IN PROGRESS***, 4 Homo sapiens 36,230 5-Jun-99
    unordered pieces.
    GB_HTG2:AC007739 158262 AC007739 Homo sapiens clone NH0091L03, ***SEQUENCING IN PROGRESS***, 4 Homo sapiens 36,230 5-Jun-99
    unordered pieces.
    GB_GSS9:AQ128685 425 AQ128685 HS_3026_B2_D10_MR CIT Approved Human Genomic Sperm Library D Homo sapiens 36,235 23-Sep-98
    Homosapiens genomic clone Plate = 3026 Col = 20 Row = H, genomic survey
    sequence.
    rxa02698 492 GB_EST18:AA704727 398 AA704727 zj21f05.s1 Soares_fetal_liver_spleen_1NFLS_S1 Homo sapiens cDNA clone Homo sapiens 40,470 24-DEC-1997
    IMAGE: 450945 3′, mRNA sequence.
    GB_PR2:AP000228 75698 AP000228 Homo sapiens genomic DNA, chromosome 21q21.2, LL56-APP region, Homo sapiens 42,616 20-Nov-99
    clone: R49K20, complete sequence.
    GB_PR2:AP000140 100000 AP000140 Homo sapiens genomic DNA, chromosome 21q21.2, LL56-APP region, clone Homo sapiens 42,616 20-Nov-99
    B2291C14-R44F3, segment 5/10, complete sequence.
    rxa02699 2271 GB_GSS12:AQ364540 497 AQ364540 nbxb0061O09r CUGI Rice BAC Library Oryza sativa genomic clone Oryza sativa 37,903 3-Feb-99
    nbxb0061O09r, genomic survey sequence.
    GB_PR4:AC006044 141509 AC006044 Homo sapiens BAC clone NH0539B24 from 7p15.1-p14, complete sequence. Homo sapiens 36,360 18-MAR-1999
    GB_PR2:HSAF001552 91526 AF001552 Homo sapiens chromosome 16 BAC clone CIT987SK-381E11 complete Homo sapiens 35,352 21-Aug-97
    sequence.
    rxa02724 967 GB_HTG2:HSDJ139D8 167079 AL096814 Homo sapiens chromosome 6 clone RP1-139D8 map p12.1-21.1, Homo sapiens 36,820 03-DEC-1999
    ***SEQUENCING IN PROGRESS***, in unordered pieces.
    GB_HTG2:HSDJ139D8 167079 AL096814 Homo sapiens chromosome 6 clone RP1-139D8 map p12.1-21.1, Homo sapiens 36,820 03-DEC-1999
    ***SEQUENCING IN PROGRESS***, in unordered pieces.
    GB_BA1:AB015853 5461 AB015853 Pseudomonas aeruginosa gene for MexX and MexY, complete cds. Pseudomonas aeruginosa 39,121 13-Nov-98
    rxa02747 2199 GB_BA1:CAJ10319 5368 AJ010319 Corynebacterium glutamicum amtP, glnB, glnD genes and partial ftsY and srp Corynebacterium glutamicum 100,000 14-MAY-1999
    genes.
    GB_GSS13:AQ463737 463 AQ463737 HS_5051_B2_D05_SP6E RPCI-11 Human Male BAC Library Homo sapiens Homo sapiens 37,549 23-Apr-99
    genomic clone Plate = 627 Col = 10 Row = H, genomic survey sequence.
    GB_BA1:CAJ10319 5368 AJ010319 Corynebacterium glutamicum amtP, glnB, glnD genes and partial ftsY and srp Corynebacterium glutamicum 100,000 14-MAY-1999
    genes.
    rxa02760 1077 GB_IN2: AC004295 84551 AC004295 Drosophila melanogaster DNA sequence (P1 DS08374 (D180)), complete Drosophila melanogaster 40,303 29-Jul-98
    sequence.
    GB_HTG6:AC011647 141830 AC011647 Homo sapiens clone RP11-15D18, ***SEQUENCING IN PROGRESS***, 29 Homo sapiens 38,158 04-DEC-1999
    unordered pieces.
    GB_HTG6:AC011647 141830 AC011647 Homo sapiens clone RP11-15D18, ***SEQUENCING IN PROGRESS***, 29 Homo sapiens 36,321 04-DEC-1999
    unordered pieces.
    rxa02787 1500 GB_BA1:MLCB1259 38807 AL023591 Mycobacterium leprae cosmid B1259. Mycobacterium leprae 57,533 27-Aug-99
    GB_BA1:MSGB937CS 38914 L78820 Mycobacterium leprae cosmid B937 DNA sequence. Mycobacterium leprae 57,600 15-Jun-96
    GB_PR4:AC006474 69718 AC006474 Homo sapiens clone DJ0669I17, complete sequence. Homo sapiens 37,246 1-Jul-99
    rxa02830 662 GB_BA1:MTCY22D7 31859 Z83866 Mycobacterium tuberculosis H37Rv complete genome; segment 133/162. Mycobacterium tuberculosis 41,527 17-Jun-98
    GB_BA1:MTCY22D7 31859 Z83866 Mycobacterium tuberculosis H37Rv complete genome; segment 133/162. Mycobacterium tuberculosis 41,223 17-Jun-98
    GB_EST12:AA276025 440 AA276025 vc30a07.r1 Barstead MPLRB1 Mus musculus cDNA clone IMAGE: 776052 5′ Mus musculus 38,746 1-Apr-97
    similar to gb: L38607 Mus musculus (MOUSE);, mRNA sequence.
    rxa02831
    rxs03200 759 GB_IN2:AE001274 268984 AE001274 Leishmania major chromosome 1, complete sequence. Leishmania major 38,575 24-MAR-1999
    GB_IN2:AE001274 268984 AE001274 Leishmania major chromosome 1, complete sequence. Leishmania major 36,772 24-MAR-1999
    GB_OM:SSIFNG 5568 X53085 S. scrofa DNA for interferon-gamma. Sus scrofa 33,515 28-Jul-95
    rxs03208 565 GB_BA1:BRLDTXR 1091 L35906 Corynebacterium glutamicum (clone pULJSX4) diphtheria toxin repressor (dtxr) Brevibacterium 99,646 06-MAR-1996
    gene, complete cds. lactofermentum
    GB_BA1:MTCY05A6 38631 Z96072 Mycobacterium tuberculosis H37Rv complete genome; segment 120/162. Mycobacterium tuberculosis 61,062 17-Jun-98
    GB_BA1:CORDTXRAA 2604 M80338 Corynebacterium diphtheriae diphtheria toxin repressor (dtxR) gene, complete Corynebacterium diphtheriae 66,372 26-Apr-93
    cds.
    rxs03219 1114 GB_HTG3:AC005769 200000 AC005769 Homo sapiens chromosome 4, ***SEQUENCING IN PROGRESS***, 5 Homo sapiens 38,613 21-Aug-99
    unordered pieces.
    GB_PR3:AF015723 33189 AF015723 Homo sapiens chromosome 21q22 cosmid clone Q4B12, complete sequence. Homo sapiens 36,866 21-Jan-98
    GB_HTG3:AC007315 159747 AC007315 Homo sapiens clone NH0189B16, ***SEQUENCING IN PROGRESS***, 3 Homo sapiens 35,005 23-Apr-99
    unordered pieces.

Claims (20)

1. An isolated nucleic acid molecule selected from the group consisting of
a) an isolated nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:65, or a complement thereof;
b) an isolated nucleic acid molecule which encodes a polypeptide comprising the amino acid sequence of SEQ ID NO:66, or a complement thereof;
c) an isolated nucleic acid molecule which encodes a naturally occurring allelic variant of a polypeptide comprising the amino acid sequence of SEQ ID NO:66, or a complement thereof;
d) an isolated nucleic acid molecule comprising a nucleotide sequence which is at least 50% identical to the entire nucleotide sequence of SEQ ID NO:65, or a complement thereof; and
e) an isolated nucleic acid molecule comprising a fragment of at least 15 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:65, or a complement thereof.
2. An isolated nucleic acid molecule comprising the nucleic acid molecule of claim 1 and a nucleotide sequence encoding a heterologous polypeptide.
3. A vector comprising the nucleic acid molecule of claim 1.
4. The vector of claim 3, which is an expression vector.
5. A host cell transfected with the expression vector of claim 4.
6. The host cell of claim 5, wherein said cell is a microorganism.
7. The host cell of claim 6, wherein said cell belongs to the genus Corynebacterium or Brevibacterium.
8. A method of producing a polypeptide comprising culturing the host cell of claim 5 in an appropriate culture medium to, thereby, produce the polypeptide.
9. A method for producing a fine chemical, comprising culturing the cell of claim 5 such that the fine chemical is produced.
10. The method of claim 9, wherein said method further comprises the step of recovering the fine chemical from said culture.
11. The method of claim 9, wherein said cell belongs to the genus Corynebacterium or Brevibacterium.
12. The method of claim 9, wherein said cell is selected from the group consisting of Corynebacterium glutamicum, Corynebacterium herculis, Corynebacterium, lilium, Corynebacterium acetoacidophilum, Corynebacterium acetoglutamicum, Corynebacterium acetophilum, Corynebacterium ammoniagenes, Corynebacterium fujiokense, Corynebacterium nitrilophilus, Brevibacterium ammoniagenes, Brevibacterium butanicum, Brevibacterium divaricatum, Brevibacterium flavum, Brevibacterium healii, Brevibacterium ketoglutamicum, Brevibacterium ketosoreductum, Brevibacterium lactofermentum, Brevibacterium linens, Brevibacterium paraffinolyticum, and those strains set forth in Table 3.
13. The method of claim 9, wherein expression of the nucleic acid molecule from said vector results in modulation of production of said fine chemical.
14. The method of claim 9, wherein said fine chemical is selected from the group consisting of organic acids, proteinogenic and nonproteinogenic amino acids, purine and pyrimidine bases, nucleosides, nucleotides, lipids, saturated and unsaturated fatty acids, diols, carbohydrates, aromatic compounds, vitamins, cofactors, polyketides, and enzymes.
15. The method of claim 9, wherein said fine chemical is an amino acid selected from the group consisting of lysine, glutamate, glutamine, alanine, aspartate, glycine, serine, threonine, methionine, cysteine, valine, leucine, isoleucine, arginine, proline, histidine, tyrosine, phenylalanine, and tryptophan.
16. An isolated polypeptide selected from the group consisting of
a) an isolated polypeptide comprising the amino acid sequence of SEQ ID NO:66;
b) an isolated polypeptide comprising a naturally occurring allelic variant of a polypeptide comprising the amino acid sequence of SEQ ID NO:66;
c) an isolated polypeptide which is encoded by a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:65;
d) an isolated polypeptide which is encoded by a nucleic acid molecule comprising a nucleotide sequence which is at least 50% identical to the entire nucleotide sequence of SEQ ID NO:65;
e) an isolated polypeptide comprising an amino acid sequence which is at least 50% identical to the entire amino acid sequence of SEQ ID NO:66; and
f) an isolated polypeptide comprising a fragment of a polypeptide comprising the amino acid sequence of SEQ ID NO:66, wherein said polypeptide fragment maintains a biological activity of the polypeptide comprising the amino sequence.
17. The isolated polypeptide of claim 16, further comprising heterologous amino acid sequences.
18. A method for diagnosing the presence or activity of Corynebacterium diphtheriae in a subject, comprising detecting the presence of at least one of the nucleic acid molecules of claim 1, thereby diagnosing the presence or activity of Corynebacterium diphtheriae in the subject.
19. A method for diagnosing the presence or activity of Corynebacterium diphtheriae in a subject, comprising detecting the presence of at least one of the polypeptide molecules of claim 16, thereby diagnosing the presence or activity of Corynebacterium diphtheriae in the subject.
20. A host cell comprising a nucleic acid molecule selected from the group consisting of
a) the nucleic acid molecule of SEQ ID NO:65, wherein the nucleic acid molecule is disrupted by at least one technique selected from the group consisting of a point mutation, a truncation, an inversion, a deletion, an addition, a substitution and homologous recombination;
b) the nucleic acid molecule of SEQ ID NO:65, wherein the nucleic acid molecule comprises one or more nucleic acid modifications as compared to the sequence of SEQ ID NO:65, wherein the modification is selected from the group consisting of a point mutation, a truncation, an inversion, a deletion, an addition and a substitution; and
c) the nucleic acid molecule of SEQ ID NO:65, wherein the regulatory region of the nucleic acid molecule is modified relative to the wild-type regulatory region of the molecule by at least one technique selected from the group consisting of a point mutation, a truncation, an inversion, a deletion, an addition, a substitution and homologous recombination.
US11/508,066 1999-06-25 2006-08-21 Corynebacterium glutamicum genes encoding regulatory proteins Abandoned US20070015252A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/508,066 US20070015252A1 (en) 1999-06-25 2006-08-21 Corynebacterium glutamicum genes encoding regulatory proteins

Applications Claiming Priority (30)

Application Number Priority Date Filing Date Title
US14103199P 1999-06-25 1999-06-25
US14269099P 1999-07-01 1999-07-01
DE19930476 1999-07-01
DE19930476.9 1999-07-01
DE19931419 1999-07-08
DE19931420.9 1999-07-08
DE19931420 1999-07-08
DE19931419.5 1999-07-08
DE19932122.1 1999-07-09
DE19932207 1999-07-09
DE19932134.5 1999-07-09
DE19932206.6 1999-07-09
DE19932122 1999-07-09
DE19932207.4 1999-07-09
DE19932134 1999-07-09
DE19932128 1999-07-09
DE19932206 1999-07-09
DE19932128.0 1999-07-09
DE19933003.4 1999-07-14
DE19933003 1999-07-14
US15125199P 1999-08-27 1999-08-27
DE19941390 1999-08-31
DE19941390.8 1999-08-31
DE19942088 1999-09-03
DE19942124.2 1999-09-03
DE19942124 1999-09-03
DE19942088.2 1999-09-03
US60287400A 2000-06-23 2000-06-23
US11/006,098 US20050153402A1 (en) 1999-06-25 2004-12-06 Corynebacterium glutamicum genes encoding regulatory proteins
US11/508,066 US20070015252A1 (en) 1999-06-25 2006-08-21 Corynebacterium glutamicum genes encoding regulatory proteins

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/006,098 Division US20050153402A1 (en) 1999-06-25 2004-12-06 Corynebacterium glutamicum genes encoding regulatory proteins

Publications (1)

Publication Number Publication Date
US20070015252A1 true US20070015252A1 (en) 2007-01-18

Family

ID=34744069

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/006,098 Abandoned US20050153402A1 (en) 1999-06-25 2004-12-06 Corynebacterium glutamicum genes encoding regulatory proteins
US11/508,066 Abandoned US20070015252A1 (en) 1999-06-25 2006-08-21 Corynebacterium glutamicum genes encoding regulatory proteins
US11/508,134 Abandoned US20070059809A1 (en) 1999-06-25 2006-08-21 Corynebacterium glutamicum genes encoding regulatory proteins

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/006,098 Abandoned US20050153402A1 (en) 1999-06-25 2004-12-06 Corynebacterium glutamicum genes encoding regulatory proteins

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/508,134 Abandoned US20070059809A1 (en) 1999-06-25 2006-08-21 Corynebacterium glutamicum genes encoding regulatory proteins

Country Status (1)

Country Link
US (3) US20050153402A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012130783A2 (en) 2011-03-27 2012-10-04 Arch Chemicals Inc. Modulating epigenetic dna methylation to cause cells to adopt dna methylation patterns associated with young cells
CN109207463A (en) * 2018-09-14 2019-01-15 宿州学院 A kind of L-Aspartic acid enzyme gene engineering bacterium fermentation culture medium and fermented and cultured L-Aspartic acid enzyme gene engineering bacteria method
EP3567110A4 (en) * 2018-01-25 2020-07-08 CJ Cheiljedang Corporation Microorganism of genus corynebacterium for producing l-amino acid and method for producing l-amino acid by using same
CN111788308A (en) * 2018-01-25 2020-10-16 Cj第一制糖株式会社 Microorganism of corynebacterium species producing purine nucleotide and method for producing purine nucleotide by using the same

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7892798B2 (en) * 1999-06-25 2011-02-22 Evonik Degussa Gmbh Nucleic acid molecules encoding metabolic regulatory proteins from Corynebacterium glutamicum, useful for increasing the production of methionone by a microorganism
US7132272B2 (en) * 1999-10-05 2006-11-07 Degussa Ag Nucleotide sequence encoding corynebacterium glutamicum leucine response regulatory protein
US6395528B1 (en) * 2000-01-27 2002-05-28 Ajinomoto Co., Inc. Phosphoserine phosphatase gene of coryneform bacteria
DE10039043A1 (en) * 2000-08-10 2002-02-21 Degussa New nucleotide sequences coding for the luxR gene
DE10359661A1 (en) * 2003-12-18 2005-07-28 Basf Ag Gene variants coding for proteins from the metabolic pathway of fine chemicals
KR101078611B1 (en) * 2005-08-26 2011-11-01 아지노모토 가부시키가이샤 L-glutamic acid-producing bacterium and method for production of L-glutamic acid
JPWO2008001597A1 (en) * 2006-06-30 2009-11-26 味の素株式会社 Protein production method
WO2008001597A1 (en) * 2006-06-30 2008-01-03 Ajinomoto Co., Inc. Method for production of protein
KR100824457B1 (en) * 2006-10-16 2008-04-22 씨제이제일제당 (주) A microorganism producing Glutamic acid in high yield and a process of producing Glutamic acid using the same
KR100838038B1 (en) * 2006-12-29 2008-06-12 씨제이제일제당 (주) - - a microorganism of corynebacterium genus having enhanced l-lysine productivity and a method of producing l-lysine using the same
KR100838035B1 (en) * 2006-12-29 2008-06-12 씨제이제일제당 (주) - - a microorganism of corynebacterium genus having enhanced l-lysine productivity and a method of producing l-lysine using the same
KR100830826B1 (en) * 2007-01-24 2008-05-19 씨제이제일제당 (주) Process for producing fermentation product from carbon sources containing glycerol using corynebacteria
US8133714B2 (en) 2007-09-27 2012-03-13 Evonik Degussa Gmbh Process for the fermentative preparation of organic chemical compounds using coryneform bacteria in which the SugR gene is present in attenuated form
KR101126041B1 (en) * 2008-04-10 2012-03-19 씨제이제일제당 (주) A transformation vector using transposon, a microorganism transformed with the vector and method of producing l-lysine using the microorganism
US8932861B2 (en) 2008-04-10 2015-01-13 Cj Cheiljedang Corporation Transformation vector comprising transposon, microorganisms transformed with the vector, and method for producing L-lysine using the microorganism
KR20140014648A (en) * 2012-07-25 2014-02-06 삼성전자주식회사 Modified microorganism for high efficient production of 1,4-butanediol
ES2758096T3 (en) * 2015-03-25 2020-05-04 Senseup Gmbh Sensors for the detection and quantification of microbiological protein secretion
KR101793328B1 (en) * 2015-07-03 2017-11-03 씨제이제일제당 (주) A microorganism having enhanced L-lysine productivity and a method of producing L-lysine using the same
KR102335301B1 (en) * 2016-10-21 2021-12-06 아지노모토 가부시키가이샤 secretory production method of protein
EP3594355A1 (en) * 2018-07-12 2020-01-15 Evonik Operations GmbH Method for the fermentative production of l-lysine
CN112143751B (en) * 2020-09-22 2022-05-06 廊坊梅花生物技术开发有限公司 Bacillus subtilis engineering bacterium for high nucleoside yield, and construction method and application thereof
KR102306009B1 (en) * 2021-04-07 2021-09-27 씨제이제일제당 (주) Novel WhiB family transcriptional regulator WhcA variant and a method for producing L-valine using the same
CN114107141B (en) * 2021-08-19 2022-07-12 中国科学院天津工业生物技术研究所 Corynebacterium glutamicum capable of producing L-proline in high yield and method for producing L-proline in high yield
CN114107148B (en) * 2021-11-12 2023-12-29 清华大学 Genetically engineered strain for high-yield hyaluronic acid and application thereof
CN115124605B (en) * 2022-03-15 2023-08-04 吉林大学 High temperature resistant element mutant and application thereof in amino acid production

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5643758A (en) * 1987-03-10 1997-07-01 New England Biolabs, Inc. Production and purification of a protein fused to a binding protein
US5912142A (en) * 1996-11-22 1999-06-15 Duke University Gene product over expressed in cancer cells

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6372453B1 (en) * 1997-02-18 2002-04-16 Genetech, Inc. Neurturin receptor

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5643758A (en) * 1987-03-10 1997-07-01 New England Biolabs, Inc. Production and purification of a protein fused to a binding protein
US5912142A (en) * 1996-11-22 1999-06-15 Duke University Gene product over expressed in cancer cells

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012130783A2 (en) 2011-03-27 2012-10-04 Arch Chemicals Inc. Modulating epigenetic dna methylation to cause cells to adopt dna methylation patterns associated with young cells
EP3567110A4 (en) * 2018-01-25 2020-07-08 CJ Cheiljedang Corporation Microorganism of genus corynebacterium for producing l-amino acid and method for producing l-amino acid by using same
CN111788308A (en) * 2018-01-25 2020-10-16 Cj第一制糖株式会社 Microorganism of corynebacterium species producing purine nucleotide and method for producing purine nucleotide by using the same
AU2019212400B2 (en) * 2018-01-25 2022-04-14 Cj Cheiljedang Corporation Microorganism Of Genus Corynebacterium For Producing L-Amino Acid And Method For Producing L-Amino Acid By Using Same
AU2019212400B8 (en) * 2018-01-25 2022-05-12 Cj Cheiljedang Corporation Microorganism Of Genus Corynebacterium For Producing L-Amino Acid And Method For Producing L-Amino Acid By Using Same
CN109207463A (en) * 2018-09-14 2019-01-15 宿州学院 A kind of L-Aspartic acid enzyme gene engineering bacterium fermentation culture medium and fermented and cultured L-Aspartic acid enzyme gene engineering bacteria method

Also Published As

Publication number Publication date
US20070059809A1 (en) 2007-03-15
US20050153402A1 (en) 2005-07-14

Similar Documents

Publication Publication Date Title
US6696561B1 (en) Corynebacterium glutamicum genes encoding proteins involved in membrane synthesis and membrane transport
US20070015252A1 (en) Corynebacterium glutamicum genes encoding regulatory proteins
US7510854B2 (en) Corynebacterium glutamicum genes encoding metabolic pathway proteins
AU783703B2 (en) Corynebacterium glutamicum genes encoding stress, resistance and tolerance proteins
US20070161091A1 (en) Corynebacterium glutamicum genes encoding proteins involved in genetic stability, gene expression, and protein secretion and folding
EP2292763A1 (en) Corynebacterium glutamicum genes encoding proteins involved in carbon metabolism and energy production
US7410766B2 (en) Corynebacterium glutamicum genes encoding phosphoenolpyruvate: sugar phosphotransferase system proteins
US20070059810A1 (en) Corynebacterium glutamicum genes encoding stress, resistance and tolerance proteins
US20050244935A1 (en) Corynebacterium glutamicum genes encoding proteins involved in membrane synthesis and membrane transport
EP1246922B1 (en) Corynebacterium glutamicum genes encoding phosphoenolpyruvate: sugar phosphotransferase system proteins
US20060269975A1 (en) Corynebacterium glutamicum genes encoding proteins involved in DNA replication, protein synthesis, and pathogenesis
US20070111232A1 (en) Corynebacterium glutamicum genes encoding proteins involved in homeostasis and adaptation
US20050191732A1 (en) Corynebacterium glutamicum genes encoding proteins involved in homeostasis and adaptation
US20080096211A1 (en) Corynebacterium glutamicum genes encoding proteins involved in genetic stability, gene expression, and protein secretion and folding
CA2585907A1 (en) Corynebacterium glutamicum genes encoding proteins involved in homeostasis and adaptation
AU783707B2 (en) Corynebacterium glutamicum genes encoding proteins involved in membrane synthesis and membrane transport
AU2007202317A1 (en) Corynebacterium glutamicum genes encoding phosphoenolpyruvate: sugar phosphotransferase system proteins
CA2583703A1 (en) Corynebacterium glutamicum genes encoding proteins involved in membrane synthesis and membrane transport
EP1702980A1 (en) Corynebacterium glutamicum gene encoding Hpr of phosphoenolpyruvate:sugar phosphotransferase system
AU2006200801A1 (en) Corynebacterium glutamicum genes encoding stress, resistance and tolerance proteins
CA2590403A1 (en) Corynebacterium glutamicum genes encoding phosphoenolpyruvate:sugar phospho-transferase system proteins
CA2585184A1 (en) Corynebacterium glutamicum genes encoding stress, resistance and tolerance proteins
AU2007202394A1 (en) Corynebacterium glutamicum genes encoding stress, resistance and tolerance proteins
EP1683859A2 (en) Putative gene coding for Catechol 1,2-Dioxygenase from Corynebacterium glutamicum
AU2006200800A1 (en) Corynebacterium Glutamicum genes encoding phosphoenolpyruvate: sugar phosphotransferase system proteins

Legal Events

Date Code Title Description
AS Assignment

Owner name: BASF AG, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:POMPEJUS, MARKUS;KROGER, BURKHARD;HABERHAUER, GREGOR;AND OTHERS;REEL/FRAME:018222/0195

Effective date: 20020409

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION